1
|
Akabane M, Imaoka Y, Kawashima J, Pawlik TM. Advancing precision medicine in hepatocellular carcinoma: current challenges and future directions in liquid biopsy, immune microenvironment, single nucleotide polymorphisms, and conversion therapy. Hepat Oncol 2025; 12:2493457. [PMID: 40260687 PMCID: PMC12026093 DOI: 10.1080/20450923.2025.2493457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a health concern characterized by heterogeneity and high mortality. Surgical resection, radiofrequency ablation, trans-arterial chemoembolization, and liver transplantation offer potentially curative treatments for early-stage disease, but recurrence remains high. Most patients present with advanced-stage HCC, where locoregional therapies are less effective, and systemic treatments-primarily multi-kinase inhibitors and immune checkpoint inhibitors-often yield limited responses. Precision medicine aims to tailor therapy to molecular and genetic profiles, yet its adoption in HCC is hindered by inter-/intra-tumoral heterogeneity and limited biopsy availability. Advances in molecular diagnostics support reintroducing tissue sampling to better characterize genetic, epigenetic, and immunological features. Liquid biopsy offers a minimally invasive method for capturing real-time tumor evolution, overcoming spatial and temporal heterogeneity. Artificial intelligence and machine learning are revolutionizing biomarker discovery, risk stratification, and treatment planning by integrating multi-omics data. Immunological factors such as tumor-infiltrating lymphocytes, natural killer cells, macrophages, and fibroblasts have emerged as determinants of HCC progression and treatment response. Conversion therapy-combining systemic agents with locoregional treatments-has showndemonstrated promise in downstaging unresectable HCC. Ongoing efforts to refine biomarker-driven approaches and optimize multi-modality regimens underscore precision medicine's potential to improve outcomes. PubMed (January 2002-February 2025) was searched for relevant studies.
Collapse
Affiliation(s)
- Miho Akabane
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Yuki Imaoka
- Division of Abdominal Transplant, Department of Surgery, Stanford University, CA, USA
| | - Jun Kawashima
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
2
|
Darbà J, Ascanio M. Hepatocellular carcinoma: what are the differential costs compared to the general population? J Med Econ 2025; 28:471-478. [PMID: 40126406 DOI: 10.1080/13696998.2025.2484073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC), which accounts for about 90% of all primary liver cancer cases, is the fifth most common malignancy and the second leading cause of cancer-related mortality worldwide. This study aims to analyse the differential costs of HCC-related hospital admissions compared to the general population in Spain. METHODS A retrospective multicenter study analyzed inpatient admissions from a Spanish national discharge database, covering 90% of hospitals between 2010 and 2022. HCC-related admissions were identified using ICD-9 and ICD-10 codes, while control admissions were selected from the general population in the same database without an HCC diagnosis. The direct hospitalization cost was included, covering medical examinations, procedures, medications, surgeries, personnel and equipment. Statistical methods, including nearest-neighbor matching, propensity score matching, and a generalized linear model, were used to estimate differential costs and to ensure comparability based on age, gender, and Charlson Comorbidity Index (CCI). RESULTS A total of 199,670 HCC-related hospital admissions and 200,000 control admissions were analyzed. Most HCC-related admissions involved male patients (78%) aged 66-85 years, with an average CCI of 5.18. HCC-related admissions incurred significantly higher costs, with an estimated differential cost of €1,303.68 using GLM, €1,804.25 via propensity score matching, and €1,767.77 using nearest-neighbor matching. Total costs per HCC admission ranged between €1,000 and €31,000. CONCLUSIONS HCC-related hospital admissions impose a significantly higher economic burden due to the complexity of care. Given the high mortality and resource utilization, advancements in early detection, treatment, and cost-effective interventions are needed to improve patient outcomes and reduce healthcare costs.
Collapse
Affiliation(s)
- Josep Darbà
- Department of Economics, Universitat de Barcelona, Barcelona, Spain
| | | |
Collapse
|
3
|
Wang Q, Sun L, Zhang G, Chen Z, Li G, Jin R. A novel nomogram based on machine learning predicting overall survival for hepatocellular carcinoma patients with dynamic α‑fetoprotein level changes after local resection. Oncol Lett 2025; 29:310. [PMID: 40342725 PMCID: PMC12059617 DOI: 10.3892/ol.2025.15056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/20/2025] [Indexed: 05/11/2025] Open
Abstract
The principal aim of the present study was to develop and validate a nomogram predicting overall survival (OS) in patients with α-fetoprotein (AFP)-negative hepatocellular carcinoma (AFP-NHCC) who experience dynamic changes in AFP level after hepatectomy. A cohort of 870 patients were enrolled and randomly assigned into a training cohort (n=600) and a validation cohort (n=270) at a 7:3 ratio. The key variables contributing to the nomogram were determined through random survival forest analysis and multivariate Cox regression. The discriminative ability of the nomogram was evaluated using time-dependent receiver operating characteristic curves and the area under the curves. Furthermore, the nomogram was comprehensively assessed using the concordance index (C-index), calibration curves and clinical decision curve analysis (DCA). Kaplan-Meier (KM) curves analysis was employed to discern survival rates across diverse risk strata of patients. Ultimately, the nomogram incorporated critical factors including sex, tumor size, globulin levels, gamma-glutamyl transferase and fibrinogen levels. In the training and validation cohorts, the C-indexes were 0.72 [95% confidence interval (CI): 0.685-0.755) and 0.664 (95% CI: 0.611-0.717], respectively, attesting to its predictive validity. The nomogram demonstrated excellent calibration and DCA further confirmed its clinical usefulness. Additionally, KM curve analysis unveiled statistically significant differences in OS among three distinct risk groups. In conclusion, the present study successfully formulated a nomogram predicting 3-, 5- and 8-year OS in patients with AFP-NHCC with dynamic changes in AFP level post-local resection. This model serves as a valuable tool for clinicians to promptly identify high-risk patients, thereby facilitating timely interventions and potentially enhancing patient survival outcomes.
Collapse
Affiliation(s)
- Qi Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
- Beijing Institute of Hepatology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Lina Sun
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
- Beijing Institute of Hepatology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Gongming Zhang
- Department of General Surgery, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Zhuangzhuang Chen
- Department of General Surgery, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Guangming Li
- Department of General Surgery, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Ronghua Jin
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
- Beijing Institute of Hepatology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
4
|
Qi M, Zhang K, Zhang X, Zhu Y, Cai B, Wang C, Zhao G, Zhang D, Zhang J. Arginine tagged liposomal carrier for the delivery of celastrol for ferroptosis-induced hepatocellular carcinoma therapy. Colloids Surf B Biointerfaces 2025; 250:114546. [PMID: 39919344 DOI: 10.1016/j.colsurfb.2025.114546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
Hepatocellular carcinoma (HCC) is a predominant malignant liver tumor that cannot be efficiently treated because of poor response, toxicity, and drug resistance. Ferroptosis is an iron-dependent way of cell death associated with abnormal intracellular lipid metabolism. Celastrol (Cel) has the ability to inhibit the progression of HCC by regulating multiple signaling pathways and induce ferroptosis. However, Cel exists the limitations of low water solubility, low oral bioavailability, and high organ toxicity. Cel was encapsulated in polyethylene glycol-based liposomes modified with L-arginine (Cel@Lip-Arg). Cel@Lip-Arg has a uniform size distribution (∼100 nm), high drug loading (80 %), and excellent ability to target liver cancer cells. In vitro experiments demonstrated that Cel@Lip-Arg considerably suppressed the activity of HuH7 (hepatoma) cells but had a negligible effect on L02 (normal) cells. Cel@Lip-Arg induced ferroptosis in hepatoma cells by promoting transferrin receptor expression, inhibiting system xc- and glutathione peroxidase 4, and favoring intracellular peroxide accumulation. In vivo experiments revealed that Cel@Lip-Arg plays a therapeutic role by inducing ferroptosis. Compared to Cel, Cel@Lip-Arg had a higher anti-hepatoma activity and effectively reduced the toxicity of Cel in mice. Cel@Lip-Arg-induced ferroptosis was concluded to be an attractive strategy for the precise treatment of HCC.
Collapse
Affiliation(s)
- Manman Qi
- School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Kai Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Xue Zhang
- School of Basic Medicine, Ningxia Medical University, Ningxia 750004, PR China
| | - Yuzhao Zhu
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China
| | - Banglan Cai
- School of Basic Medicine, Ningxia Medical University, Ningxia 750004, PR China
| | - Chao Wang
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China
| | - Gang Zhao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Denghai Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China.
| | - Jian Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China; Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China.
| |
Collapse
|
5
|
Fu L, Gu X, Lou N, Li J, Xue C. Current research of the Notch pathway in hepatocellular carcinoma. Eur J Med Res 2025; 30:402. [PMID: 40394648 PMCID: PMC12090635 DOI: 10.1186/s40001-025-02626-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025] Open
Abstract
Notch signaling is a widely preserved communication pathway that supports essential cellular functions by allowing adjacent cells to interact. The Notch signaling pathway consists of Notch ligands (DSL proteins), Notch receptors, DNA-binding proteins, and downstream target genes. Hepatocellular carcinoma (HCC) represents the predominant cause of cancer-related deaths globally and poses a significant threat to human health. For highly malignant HCC, current treatment options, including chemotherapy, radiotherapy, immunotherapy, targeted therapies, and surgical procedures, often have poor prognoses. Therefore, there is a need to explore additional therapeutic strategies. Many studies have found that abnormal activation of the Notch signaling pathway contributes to tumor initiation and progression by promoting HCC proliferation, metastasis, stem cell-like properties, and drug resistance. In this research, we reveal the composition and activation mechanisms of the Notch signaling pathway, as well as the molecular mechanism underlying its aberrant activation in HCC. Furthermore, we summarize recent advances in targeting Notch signaling for the treatment of HCC. This review aims to highlight the promising potential of investigating the Notch pathway as a therapeutic target in HCC.
Collapse
Affiliation(s)
- Leiya Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Na Lou
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
6
|
Yin M, Wang L, Liu Y, Chen J, Gao H, Xu J, Guo Y, Cui X, Yu G, Cai C. GSH-Responsive GalNAc-Conjugated Glycopolymer for Targeted Survivin siRNA Delivery in Hepatocellular Carcinoma Therapy. ACS Macro Lett 2025; 14:589-596. [PMID: 40269699 DOI: 10.1021/acsmacrolett.5c00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Gene interference therapy has made significant progress in the treatment of various diseases by targeting specific pathogenic genes and down-regulating the production of harmful proteins. This approach enables the precise modulation of gene expression, offering potential therapeutic benefits for conditions driven by genetic mutations or abnormal protein accumulation. Survivin, an apoptosis-inhibiting protein, plays a critical role in regulating tumor cell proliferation and preventing programmed cell death. Its overexpression in liver cancer cells is strongly associated with poor prognosis and accelerated tumor progression. RNA interference (RNAi) therapy can effectively suppress the expression of Survivin in liver cancer, inhibiting tumor cell proliferation and promoting apoptosis. In this study, four distinct GalNAc-conjugated glycopolymer siRNA delivery systems were developed. By leveraging the efficient liver-targeting capability of the GalNAc moiety, Survivin-siRNA was specifically delivered to liver cancer cells through either covalent coupling or electrostatic adsorption. In vitro experiments demonstrated the excellent gene silencing effect of these siRNA complexes, highlighting their potential as a promising therapeutic strategy for liver cancer.
Collapse
Affiliation(s)
- Mengfei Yin
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
| | - Lihao Wang
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
| | - Yang Liu
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
| | - Jingjing Chen
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
| | - Hongming Gao
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
| | - Jinlong Xu
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
| | - Yuxin Guo
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
| | - Xinying Cui
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
| | - Guangli Yu
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Chao Cai
- Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
7
|
Gao Y, Chen J, Du W. Identification of novel potential biomarkers using bulk RNA and single cells to build a neural network model for diagnosis of liver cancer. Discov Oncol 2025; 16:728. [PMID: 40353917 PMCID: PMC12069198 DOI: 10.1007/s12672-025-02420-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND As a common cancer, liver cancer imposes an unacceptable burden on patients, but its underlying molecular mechanisms are still not fully understood. Therefore, there is an urgent need to potential biomarkers and diagnostic models for liver cancer. METHODS In this study, transcriptome and single-cell datasets related to liver cancer were downloaded from the UCSC Xena database and the Mendeley database, and differential analysis and weighted gene co-expression network analysis were used to find differentially expressed genes related to liver cancer. We used multiple machine algorithms to find hub genes related to liver cancer, and constructed new artificial neural network models based on their transcriptome expression patterns to assist in the diagnosis of liver cancer. Subsequently, we conducted survival analysis and immune infiltration analysis to explore the correlation between hub genes and immune cells, and used single-cell data to verify hub genes related to liver cancer. RESULTS This study identified MARCO, KCNN2, NTS, TERT and SFRP4 as central genes associated with liver cancer, and constructed a new artificial neural network model for molecular diagnosis of liver cancer. The diagnostic performance of the training cohort and the validation cohort was good, with the areas under the ROC curves of 1.000 and 0.986, respectively. Immune infiltration analysis determined that these central genes were closely associated with different types of immune cells. The results of immunohistochemistry and the results at the single cell level were consistent with those at the transcriptome level, and also showed obvious differences between different cell types in liver cancer and healthy states. CONCLUSION This study identified MARCO, KCNN2, NTS, TERT, and SFRP4 from multiple dimensions and highlighted their key roles in the diagnosis and treatment of liver cancer from multiple dimensions, providing promising biomarkers for the diagnosis of liver cancer.
Collapse
Affiliation(s)
- Yingzheng Gao
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Jiahao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Weidong Du
- The First Affiliated Hospital of Zhejiang, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Chinese Medical University, Hangzhou, 310006, China.
| |
Collapse
|
8
|
Zhang L, Wang H, Liang B, Qin L, Zhang M, Lv X, Hu S, Fan X, Xie W, Yang H, Huang G, Jing W, Zhao J. Ponicidin promotes ferroptosis to enhance treatment sensitivity in Lenvatinib-resistant hepatocellular carcinoma cells through regulation of KEAP1/NRF2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156824. [PMID: 40382941 DOI: 10.1016/j.phymed.2025.156824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/17/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE This study explores the therapeutic potential of Ponicidin on Lenvatinib-resistant hepatocellular carcinoma (HCC), elucidates its mechanism in reversing Lenvatinib resistance, and provides experimental evidence for its clinical application in overcoming this resistance. METHODS Huh7 and HCC-LM3 cells were used to construct Lenvatinib-resistant cell lines, Huh7-LR and HCC-LM3-LR. Changes in the ferroptosis pathway post-drug resistance were observed by measuring ferroptosis-related markers. The proliferation assay were assessed by CCK-8, while the migration and invasion were measured by scratch and Transwell invasion assays. In mechanistic study, chip analysis and immunoprecipitation with biotin-labeled Ponicidin, were conducted to explore how Ponicidin overcame drug resistance. Xenograft model in nude mice was established to examine Ponicidin's anti-HCC effects In vivo. Clinical specimens were used to assess the true status of patients in Lenvatinib-resistant HCC patients. RESULTS Our study reveals for the first time that ferroptosis inhibition drives Lenvatinib resistance in HCC and identifies Ponicidin as a novel KEAP1-targeting agent to reverse this process. In vitro, ferroptosis pathway was suppressed in Lenvatinib-resistant cells. Ponicidin suppressed proliferation, clonogenicity, migration, and invasion in these cells. The combination of Ponicidin and Lenvatinib significantly inhibited proliferation and reversed drug resistance by activating the ferroptosis pathway. Preliminary mechanistic studies showed that Ponicidin binds to KEAP1, stabilizing the KEAP1/NRF2 interaction, inhibiting the nuclear translocation and activation of NRF2, and thereby inducing ferroptosis to overcome Lenvatinib resistance. In vivo, the combination of Ponicidin and Lenvatinib exhibited a synergistic effect, significantly delaying tumor growth. Clinically, p-NRF2 and GPX4 expression was higher in the Lenvatinib-insensitive group, suggesting that the ferroptosis pathway was inhibited in these patients. Thus, this study demonstrated that Ponicidin promotes ferroptosis to enhances treatment sensitivity in Lenvatinib-resistant HCC cells through KEAP1/NRF2.
Collapse
Affiliation(s)
- Lisha Zhang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; Nanchang Hongdu Hospital of TCM, Nanchang, PR China
| | - Hao Wang
- Department of Oncology, The Air Force Hosptital of Northern Theater PLA, Shenyang, China
| | - Beibei Liang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Lijuan Qin
- Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Mingzhu Zhang
- University of Shanghai for Science and Technology, Shanghai, PR China
| | - Xingxian Lv
- Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Shi Hu
- Department of Biomedical Engineering, College of Basic Medical Sciences, Second Military Medical University, Shanghai, PR China
| | - Xiaoyu Fan
- Department of Molecular Biology, Shanghai Center for Clinical Laboratory, Shanghai, PR China
| | - Wei Xie
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China
| | - Wei Jing
- Department of Surgery, Changhai Hospital, Second Military Medical University, Shanghai, PR China.
| | - Jian Zhao
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, PR China.
| |
Collapse
|
9
|
Tian Y, Ma L, Liu S, Bai X, Shah N, Zhang L, Wang X, Zhang Y, Ding X. The prognostic value of AST-lymphocyte ratio index in liver cancer patients treated with TACE: a systematic review and single-center retrospective study. BMC Gastroenterol 2025; 25:348. [PMID: 40340835 PMCID: PMC12063415 DOI: 10.1186/s12876-025-03949-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND AND AIMS AST-lymphocyte ratio index (ALRI) has been proposed as a potentially prognostic indicator of liver cancer patients underwent transcatheter arterial chemoembolization (TACE) in studies, but the numbers were small and the results were controversial. In this study, we systematically assessed the prognostic value of ALRI in liver cancer patients treated with TACE by integrating meta-analysis with single-center clinical analysis. METHODS We conducted a systematic literature search across multiple databases and evaluated the quality of included studies using the Newcastle-Ottawa Scale. We employed a fixed-effect model to calculate the pooled hazard ratio (HR) and 95% confidence interval (CI). Publication bias were evaluated using funnel plot, Begg's and Egger's tests. Concurrently, we integrated clinical data from 127 HCC patients treated with TACE at our center, employed X-tile software to ascertain the optimal cutoff value for ALRI, and analyzed the relationship between ALRI and clinical characteristics as well as overall survival (OS), using chi-square tests, Kaplan-Meier survival curves, and Cox proportional hazards models. RESULTS The meta-analysis included 7 studies, and the pooled hazard ratio (HR) indicated that elevated ALRI was significantly associated with poorer OS in liver cancer patients underwent TACE (HR = 1.75, 95% CI: 1.46-2.1, P<0.01), with no significant heterogeneity (P = 0.542, I2 = 0.00%). Clinical analysis of 127 patients further supported this finding, with patients in the high ALRI group showed significantly lower OS compared to those in the low ALRI group (1-year OS rate: 96.7% vs. 87.9%, 2-year OS rate: 61.5% vs. 42.7%; C2 = 28.006, P<0.01). Multivariate Cox regression analysis revealed that number of tumors, tumor size and ALRI were all independent prognostic factors for OS (ALRI HR = 6.456, 95%CI: 2.247-18.55, P < 0.01). CONCLUSIONS An increase in ALRI may serve as an independent prognostic indicator of poor outcomes in liver cancer patients undergoing TACE. While it offers benefits such as being non-invasive and cost-effective, further large-scale, multicenter, prospective studies are essential to validate the efficacy of ALRI and establish standardized cutoff values for clinical application.
Collapse
Affiliation(s)
- Yali Tian
- Department of Infectious Disease, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, No. 301, Zhengyuan North Street, Jinfeng District, Yinchuan, Ningxia Hui Autonomous Region, 750021, China
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Lina Ma
- Department of Infectious Disease, General Hospital of Ningxia Medical University, No. 804, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Shuaiwei Liu
- Department of Infectious Disease, General Hospital of Ningxia Medical University, No. 804, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Xiaoyang Bai
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Nawaz Shah
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Le Zhang
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Xia Wang
- Ningxia Medical University, No. 692, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Yuxi Zhang
- Department of Infectious Disease, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, No. 301, Zhengyuan North Street, Jinfeng District, Yinchuan, Ningxia Hui Autonomous Region, 750021, China.
| | - Xiangchun Ding
- Department of Infectious Disease, General Hospital of Ningxia Medical University, No. 804, Shengli Street, Xingqing District, Yinchuan, Ningxia Hui Autonomous Region, 750004, China.
- Infectious Disease Clinical Research Center of Ningxia, 804 Shengli Street, Xingqing District, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
10
|
Muthusamy G, Liu CC, Johnston AN. LFHP-1c Attenuates Hepatocellular Carcinoma Viability In Vitro Independent of PGAM5. Cancers (Basel) 2025; 17:1573. [PMID: 40361499 PMCID: PMC12071907 DOI: 10.3390/cancers17091573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/12/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Upregulation of phosphoglycerate mutase 5 (PGAM5) is correlated with reduced survival outcomes in hepatocellular carcinoma (HCC). PGAM5 knockdown or knockout attenuates HCC growth in in vitro and in vivo models. A novel small molecule inhibitor of PGAM5, LFHP-1c, has recently been characterized. The objective of this study was to determine if LFHP-1c effectively reduces HCC viability in cell models. METHODS The hepatoma and HCC cell lines, HepG2 and HuH7, respectively, were treated with LFHP-1c. Label-free imaging was used to quantify growth. Cellular viability and reactive oxygen species (ROS) production were measured using luminescent or fluorescent assays. Expression of antioxidant and metabolic proteins was measured by immunoblot. HepG2 and HuH7 PGAM5 knockout cell lines were used as negative controls. RESULTS Treatment with LFHP-1c reduced cell growth and viability in HepG2 and HuH7 cell lines. Reactive oxygen species production was upregulated in both wild-type and PGAM5 knockout cell lines following LFHP-1c exposure. Cell viability was reduced following LFHP-1c treatment in PGAM5 knockout cell lines. CONCLUSIONS LFHP-1c reduces hepatoma and HCC viability and enhances ROS production, but these effects are independent of PGAM5.
Collapse
Affiliation(s)
- Ganesan Muthusamy
- School of Veterinary Medicine, Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, LA 70802, USA
| | - Chin-Chi Liu
- School of Veterinary Medicine, Office of Research and Graduate Education, Louisiana State University, Baton Rouge, LA 70802, USA;
| | - Andrea N. Johnston
- School of Veterinary Medicine, Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, LA 70802, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
11
|
Zhi Y, Qiaoyun T. Screening of pivotal oncogenes modulated by DNA methylation in hepatocellular carcinoma and identification of atractylenolide I as an anti-cancer drug. Hum Cell 2025; 38:97. [PMID: 40325252 DOI: 10.1007/s13577-025-01224-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
This study was performed to identify crucial oncogenes modulated by DNA methylation in hepatocellular carcinoma (HCC) and look for new drugs for HCC treatment. The data of TCGA-LIHC cohort were obtained from UCSC database. Weighted gene co-expression network analysis and multiple machine learning algorithms were applied to screen the crucial prognosis-related genes in HCC. Then these genes were further screened by DNA methylation status. Ten-eleven translocation 1 (TET1) was overexpressed in HCC cell lines, and its biological functions and regulatory effects on the oncogenes were explored by qPCR, methylation-specific polymerase chain reaction, cell viability assay, Western blot, etc. Molecular docking was applied to evaluate the binding affinity between atractylenolide I (AT-I) and TET1, and the tumor-suppressive functions of AT-I were examined with both in vitro and in vivo models. In this work, 12 crucial genes related to HCC prognosis were obtained, among which six genes were with differential methylation status in HCC tissues, including AKR1B10, ALPK3, NQO1, NT5DC2, SFN, and SPP1. The expression levels of ALPK3 and NT5DC2 were positively regulated by TET1, the crucial mediator of demethylation. TET1 overexpression increased the viability and stemness of HCC cells. AT-I had good binding affinity with TET1, and repressed its activity. AT-I promoted the methylation of ALPK3 and NT5DC2 promoter regions, and reduced their expression, and repressed the growth of HCC cells. In summary, DNA methylation contributes to HCC progression, and AT-I represses the malignancy of HCC cells by inhibiting TET1-mediated abnormal DNA demethylation.
Collapse
Affiliation(s)
- Yang Zhi
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Digestive Disease, Affiliated Central People's Hospital of China Three Gorges University, China Three Gorges University, Yichang, China
- Department of Gastroenterology, Yichang Central People's Hospital, Yichang, China
| | - Tong Qiaoyun
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China.
- Institute of Digestive Disease, Affiliated Central People's Hospital of China Three Gorges University, China Three Gorges University, Yichang, China.
- Department of Gastroenterology, Yichang Central People's Hospital, Yichang, China.
| |
Collapse
|
12
|
Gao S, Fan L, Wang H, Wang A, Hu M, Zhang L, Sun G. NCOA5 induces sorafenib resistance in hepatocellular carcinoma by inhibiting ferroptosis. Cell Death Discov 2025; 11:215. [PMID: 40316542 PMCID: PMC12052255 DOI: 10.1038/s41420-025-02473-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 05/04/2025] Open
Abstract
NCOA5 has been identified as a crucial factor in the progression of hepatocellular carcinoma (HCC). This study investigates the expression of NCOA5 in HCC, revealing its significant overexpression in tumor tissues compared to healthy liver tissues, as evidenced by analysis of the TCGA dataset and RT-qPCR in patient samples. Higher NCOA5 levels correlate with poor overall survival, highlighting its role as a prognostic indicator. Furthermore, our findings suggest that elevated NCOA5 is associated with resistance to sorafenib, a common chemotherapeutic agent for HCC, as shown through analysis of publicly available datasets and the establishment of sorafenib-resistant HCC cell lines. Mechanistically, NCOA5 appears to inhibit ferroptosis in HCC cells by modulating glutathione peroxidase 4 (GPX4) levels. Knockdown of NCOA5 sensitizes resistant cell lines to sorafenib and induces ferroptosis by decreasing GPX4 expression. Additionally, NCOA5 regulation of GPX4 is mediated through the transcription factor MYC. In vivo studies further validate that targeting NCOA5 enhances the efficacy of sorafenib in resistant HCC models by promoting ferroptosis. Collectively, these findings underscore the potential of NCOA5 as a therapeutic target to overcome drug resistance in HCC, providing insights into its role in modulating treatment responses and patient prognosis.
Collapse
Affiliation(s)
- Shuang Gao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Lulu Fan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Huiyan Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Anqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Mengyao Hu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Lei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, 233080, China.
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China.
| |
Collapse
|
13
|
Li X, Wang Y, Liu J, Gao T, Cao L, Yan M, Li N. Dysregulation of the SREBP pathway is associated with poor prognosis and serves as a potential biomarker for the diagnosis of hepatocellular carcinoma. Mol Med Rep 2025; 31:112. [PMID: 40017126 PMCID: PMC11894594 DOI: 10.3892/mmr.2025.13477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 12/20/2024] [Indexed: 03/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a severe disease associated with a poor prognosis. The role of aberrant lipid metabolism in the development and progression of HCC necessitates detailed characterization. Sterol regulatory element‑binding proteins (SREBPs), pivotal transcription factors governing lipogenesis, are central to this process. The present study aimed to assess the regulation of HCC by the SREBP signaling pathway, examining the expression levels of genes in this pathway, the clinical implications and its prognostic value using the Kaplan‑Meier method. Pearson's correlation coefficient was used to identify the co‑expression of SREBP pathway genes in HCC. Genomic analysis examined the frequency of TP53 mutations in groups with and without SREBP pathway alterations. In addition, small interfering RNAs targeting genes of the SREBP pathway were transfected into Huh‑7 and HCC‑LM3 cell lines. Subsequently, Cell Counting Kit‑8 and Transwell assays were carried out to evaluate the viability and invasion of these cells. Reverse transcription‑quantitative PCR and western blotting were performed to investigate the expression of TP53 in response to silencing of SREBP pathway genes. Dysregulation of SREBP pathway genes was detected in HCC tissues compared with in normal liver tissues, and predicted a poor prognosis. Silencing these genes reduced the viability and invasion of HCC cells. Furthermore, abnormal SREBP pathway gene expression was associated with poor survival rates, vascular invasion, advanced tumor stage and an increased incidence of TP53 mutations. By contrast, knockdown of SREBP pathway genes decreased mutant TP53 expression at both the mRNA and protein levels in HCC cells. The findings of the present study suggested that SREBP pathway genes could serve as promising prognostic biomarkers for HCC. The combined analysis of individual gene expression levels offers offer novel insights into the pathogenesis and progression of HCC.
Collapse
Affiliation(s)
- Xiaodan Li
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
- Scientific Research Department, Shanghai University of Medicine amd Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Yuhan Wang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Junchi Liu
- Scientific Research Department, Shanghai University of Medicine amd Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Tianmiao Gao
- Scientific Research Department, Shanghai University of Medicine amd Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Lizhi Cao
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Meng Yan
- Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Na Li
- Scientific Research Department, Shanghai University of Medicine amd Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
14
|
Rafati I, Destrempes F, Yazdani L, Barat M, Karam E, Fohlen A, Nguyen BN, Castel H, Tang A, Cloutier G. Enhancing Liver Nodule Visibility and Diagnostic Classification Using Ultrasound Local Attenuation Coefficient Slope Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:807-814. [PMID: 39890529 DOI: 10.1016/j.ultrasmedbio.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/03/2025] [Accepted: 01/12/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVE B-mode ultrasound (US) presents challenges in accurately detecting and distinguishing between benign and malignant liver nodules. This study utilized quantitative US local attenuation coefficient slope (LACS) imaging to address these limitations. MATERIALS AND METHODS This is a prospective, cross-sectional study in adult patients with definable solid liver nodules at US conducted from March 2021 to December 2023. The composite reference standard included histopathology when available or magnetic resonance imaging. LACS images were obtained using a phantom-free method. Nodule visibility was assessed by computing the contrast-to-noise ratio (CNR). Classification accuracy for differentiating benign and malignant lesions was assessed with the area under the receiver operating characteristic curve (AUC), along with sensitivity and specificity. RESULTS The study enrolled 97 patients (age: 62 y ± 13 [standard deviation]), with 57.0% malignant and 43.0% benign observations (size: 26.3 ± 18.9 mm). LACS images demonstrated higher CNR (12.3 dB) compared to B-mode (p < 0.0001). The AUC for differentiating nodules and liver parenchyma was 0.85 (95% confidence interval [CI]: 0.79-0.90), with higher values for malignant (0.93, CI: 0.88-0.97) than benign nodules (0.76, CI: 0.66-0.87). A LACS threshold of 0.94 dB/cm/MHz provided a sensitivity of 0.83 (CI: 0.74-0.89) and a specificity of 0.82 (CI: 0.73-0.88). LACS mean values were higher (p < 0.0001) in malignant (1.28 ± 0.27 dB/cm/MHz) than benign nodules (0.98 ± 0.19 dB/cm/MHz). CONCLUSION LACS imaging improves nodule visibility and provides better differentiation between benign and malignant liver nodules, showing promise as a diagnostic tool.
Collapse
Affiliation(s)
- Iman Rafati
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada; Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada
| | - François Destrempes
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada
| | - Ladan Yazdani
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada; Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada
| | - Maxime Barat
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Elige Karam
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Audrey Fohlen
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Bich N Nguyen
- Department of Pathology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Hélène Castel
- Departments of Hepatology and Liver Transplantation, University of Montreal Hospital, Montréal, Québec, Canada
| | - An Tang
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada; Department of Radiology, Radiation Oncology and Nuclear Medicine, University of Montreal, Montréal, Québec, Canada; Laboratory of Clinical Image Processing, University of Montreal Hospital Research Center, Montréal, Québec, Canada.
| | - Guy Cloutier
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada; Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada; Department of Radiology, Radiation Oncology and Nuclear Medicine, University of Montreal, Montréal, Québec, Canada.
| |
Collapse
|
15
|
Yang N, Ma Z, Zhang L, Ji W, Xi Q, Li M, Jin L. Radiomics-based automated machine learning for differentiating focal liver lesions on unenhanced computed tomography. Abdom Radiol (NY) 2025; 50:2126-2139. [PMID: 39572431 PMCID: PMC11992001 DOI: 10.1007/s00261-024-04685-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 04/12/2025]
Abstract
BACKGROUND & AIMS Enhanced computed tomography (CT) is the primary method for focal liver lesion diagnosis. We aimed to use automated machine learning (AutoML) algorithms to differentiate between benign and malignant focal liver lesions on the basis of radiomics from unenhanced CT images. METHODS We enrolled 260 patients from 2 medical centers who underwent CT examinations between January 2017 and March 2023. This included 60 cases of hepatic malignancies, 93 cases of hepatic hemangiomas, 48 cases of hepatic abscesses, and 84 cases of hepatic cysts. The Pyradiomics method was used to extract radiomics features from unenhanced CT images. By using the mljar-supervised (MLJAR) AutoML framework, clinical, radiomics, and fusion models combining clinical and radiomics features were established. RESULTS In the training and validation sets, the area under the curve (AUC) values for the clinical, radiomics, and fusion models exceeded 0.900. In the external testing set, the respective AUC values for the clinical, radiomics, and fusion models were as follows: 0.88, 1.00, and 1.00 for hepatic cysts; 0.81, 0.90, and 0.97 for hepatic hemangiomas; 0.89, 0.98, and 0.92 for hepatic abscesses; and 0.23, 0.80, and 0.93 for hepatic malignancies. The diagnostic accuracy rates for hepatic cysts, hemangiomas, malignancies, and abscesses by radiologists in the external testing cohort were 0.96, 0.60, 0.79, and 0.66, respectively. CONCLUSION The fusion model based on noninvasive radiomics and clinical features of unenhanced CT images has high clinical value for distinguishing focal hepatic lesions.
Collapse
Affiliation(s)
- Nan Yang
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Zhuangxuan Ma
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Ling Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wenbin Ji
- Radiology Department, Shanghai Electric Power Hospital, Shanghai, China
| | - Qian Xi
- Department of Radiology, Eye & ENT Hospital of Fudan University, Shanghai, China.
| | - Ming Li
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China.
| | - Liang Jin
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Zhang J, Guo J, Qian Y, Yu L, Ma J, Gu B, Tang W, Li Y, Li H, Wu W. Quercetin Induces Apoptosis Through Downregulating P4HA2 and Inhibiting the PI3K/Akt/mTOR Axis in Hepatocellular Carcinoma Cells: An In Vitro Study. Cancer Rep (Hoboken) 2025; 8:e70220. [PMID: 40347062 PMCID: PMC12065022 DOI: 10.1002/cnr2.70220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 03/25/2025] [Accepted: 04/22/2025] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND Quercetin is a natural product with multiple activities, which possesses a promising antitumor effect on malignancies. The involvement of proline 4-hydroxylase II (P4HA2) in collagen synthesis is crucial in the growth of tumor cells. Apoptosis is a programmed cell death requisite for the stability of the intracellular environment. However, the relationship between quercetin and cell apoptosis, as well as the impact of P4HA2 in this connection, has not yet been specified in hepatocellular carcinoma(HCC). AIMS The present study used HCC cells to investigate how quercetin regulates P4HA2 and influences cell proliferation and apoptosis. METHODS AND RESULTS The outcomes reveal that quercetin can impede the viability and growth of HCC cells and generate cell apoptosis in a dose-dependent manner. Additionally, quercetin prompts downregulation of P4HA2, leading to cell apoptosis in HCC cells, and knocking down P4HA2 can enhance this effect. Furthermore, we pretreated HCC cells with inhibitors (Z-VAD-FMK, LY294002) or activators (740Y-P) and found that the PI3K/Akt/mTOR pathway was occupied with quercetin-induced cell apoptosis. CONCLUSION This investigation reveals that quercetin compels apoptosis in HCC cells by diminishing P4HA2 and restraining the PI3K/Akt/mTOR axis.
Collapse
Affiliation(s)
- Junli Zhang
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and TreatmentBengbuChina
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Jiayi Guo
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Ying Qian
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Lianchen Yu
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Junrao Ma
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Biao Gu
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
| | - Weichun Tang
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and TreatmentBengbuChina
| | - Yi Li
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
| | - Hongwei Li
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
| | - Wenjuan Wu
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
- Department of Biochemistry and Molecular BiologySchool of Laboratory Medicine, Bengbu Medical UniversityBengbuChina
| |
Collapse
|
17
|
Qiu Y, Xu J, Liao W, Yang S, Wen Y, Farag MA, Zheng L, Zhao C. Ulvan derived from Ulva lactuca suppresses hepatocellular carcinoma cell proliferation through miR-542-3p-mediated downregulation of SLC35F6. Int J Biol Macromol 2025; 308:142252. [PMID: 40118430 DOI: 10.1016/j.ijbiomac.2025.142252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025]
Abstract
Hepatocellular carcinoma (HCC) therapy still presents significant challenges, with a critical need for novel molecular targets and effective natural compound-based therapies. Despite its known oncogenic potential in other cancers, the role of SLC35F6 in HCC has not been previously reported, leaving a gap in our understanding of its function and therapeutic relevance. Here, we demonstrate that SLC35F6 is overexpressed in HCC and is associated with poor prognosis. Ulva lactuca polysaccharide (ULP), a natural extract with known antitumor properties, exerts its effects by upregulating miR-542-3p, which in turn inhibits SLC35F6 expression and significantly increases TP53 protein levels. Furthermore, TP53 is positively regulated by miR-542-3p, and our results indicate that SLC35F6 is a target gene of miR-542-3p. Knockdown of SLC35F6 in H22 and HepG2 cells markedly reduced cell growth while elevating TP53 expression, supporting SLC35F6 as a key regulatory factor in the miR-542-3p/TP53 axis. While this study did not confirm direct mutual regulation between SLC35F6 and TP53, our findings provide evidence that targeting SLC35F6 can suppress HCC progression. Collectively, these results identify SLC35F6 as a potential therapeutic target for HCC and provide mechanistic insights into its regulation through the miR-542-3p/SLC35F6/TP53 axis.
Collapse
Affiliation(s)
- Yinghui Qiu
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; School of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
| | - Jingxiang Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Wei Liao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Shuxin Yang
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yuxi Wen
- University of Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, E32004 Ourense, Spain
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Lingjun Zheng
- School of Agriculture and Biology, Shanghai JiaoTong University, Shanghai 200240, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
18
|
Yi F, Long S, Yao Y, Fu K. A Novel Signature Composed of Hypoxia, Glycolysis, Lactylation Related Genes to Predict Prognosis and Immunotherapy in Hepatocellular Carcinoma. FRONT BIOSCI-LANDMRK 2025; 30:33422. [PMID: 40302343 DOI: 10.31083/fbl33422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. The hypoxic microenvironment in HCC enhances glycolysis and co-directed lactate accumulation, which leads to increased lactylation. However, the exact biological pattern remains to be elucidated. Therefore, we sought to identify hypoxia-glycolysis-lactylation (HGL) prognosis-related signatures and validate this in vitro. METHODS Transcriptomic data of patients with HCC were collected from The Cancer Genome Atlas (TCGA), International Cancer Genome Consortium (ICGC), and Gene Expression Omnibus (GEO) databases. Differentially expressed HGL genes between HCC and normal tissues were obtained by DEseq2. The consensus clustering algorithm was employed to stratify patients into two distinct clusters. Subsequently, the single sample Gene Set Enrichment Analysis (ssGSEA), Tumor Immune Estimation Resource (TIMER) and Tumor Immune Dysfunction and Exclusion (TIDE) algorithms were utilized to assess immune infiltration and immune evasion. Least Absolute Shrinkage and Selection Operator (LASSO) and COX regression analysis were used to identify an HGL prognosis-related signature. Based on spatial transcriptome and histological data, we analyzed the expression of these genes in HCC and explored the function of Homer Scaffold Protein 1 (HOMER1) in HCC cells. RESULTS We identified 72 differentially expressed HGL genes and two HGL clusters. Cluster2, with better survival (p < 0.001), was significantly enriched in metabolic-related pathways. The HGL prognosis-related signature exhibited great predictive efficacy for patients in TCGA, ICGC, and GSE148355 databases (3-year area under the curve (AUC) = 0.822, 0.738, and 0.707, respectively). The elevated expression of HOMER1 in HCC was revealed by the combination of spatial transcriptome and histological data. Knocking down HOMER1 significantly inhibited the malignant progression of HCC cells. CONCLUSIONS We identified a signature with great predictive efficacy and discovered a gene, HOMER1, that influences the malignant progression of HCC with the potential to become a novel therapeutic target.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/mortality
- Liver Neoplasms/genetics
- Liver Neoplasms/therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/immunology
- Liver Neoplasms/mortality
- Prognosis
- Glycolysis/genetics
- Immunotherapy
- Gene Expression Regulation, Neoplastic
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Tumor Microenvironment/genetics
- Transcriptome
- Gene Expression Profiling
- Cell Line, Tumor
- Female
- Male
Collapse
Affiliation(s)
- Feng Yi
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 410083 Changsha, Hunan, China
| | - Shichao Long
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 410083 Changsha, Hunan, China
- Department of Radiology, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410083 Changsha, Hunan, China
| | - Yuanbing Yao
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 410083 Changsha, Hunan, China
| | - Kai Fu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 410083 Changsha, Hunan, China
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, 410083 Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, 410114 Changsha, Hunan, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, 410083 Changsha, Hunan, China
| |
Collapse
|
19
|
Liu H, Ge W, Yu X, Luo J, Zhang J, Yang M, Cao L, Zhang Y, Wang R, Yang C, Li P, Tian M, Peng X, Peng L, Wu D, Liu M, Liang Q, Zhang S, Li W, Rong P, Li H, Ma X, Wang W. CRISPR/Cas9-mediated SHP-1-knockout T cells combined with simvastatin enhances anti-tumor activity in humanized-PDX HCC model. iScience 2025; 28:112266. [PMID: 40241752 PMCID: PMC12003012 DOI: 10.1016/j.isci.2025.112266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/04/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) resists immunotherapy due to its immunosuppressive microenvironment. Sarcoma homology 2 domain-containing protein tyrosine phosphatase-1 (SHP-1) inhibits T cell receptor signaling, and its pharmacological inhibition is limited by poor selectivity and membrane permeability. Here, we generated CRISPR-edited SHP-1-knockout (KO) CD8+ T cells to enhance adoptive therapy against HCC. Single-cell RNA sequencing of HCC patient T cells revealed elevated SHP-1 in exhausted subsets. SHP-1-KO T cells exhibited increased effector memory T cells (TEM) proportions and enhanced IFN-γ/Granzyme B/perforin secretion, improving cytotoxicity against HCC lines. In humanized PDX models, SHP-1-KO T cells demonstrated superior tumor-killing activity. Transcriptomics identified upregulated lipid metabolism pathways, with HMGCR as a hub gene. Combining SHP-1-KO T cells with simvastatin (HMGCR inhibitor) synergistically amplified anti-HCC efficacy. This study proposes a dual strategy combining SHP-1-targeted cell therapy and metabolic modulation to overcome immunotherapy resistance, offering a translatable approach for HCC treatment.
Collapse
Affiliation(s)
- Huaping Liu
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Wu Ge
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Xiaoping Yu
- Department of Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jianwei Luo
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Juan Zhang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Min Yang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Lu Cao
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Yangnan Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ruike Wang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cejun Yang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Pei Li
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Mengyu Tian
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - XiaoPei Peng
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Lei Peng
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Di Wu
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Muqi Liu
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Qi Liang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shengwang Zhang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei Li
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Pengfei Rong
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| | - Hailan Li
- Department of Radiology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University
| | - Xiaoqian Ma
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
Ran Y, Li L, Wang Z, Sun T, Wen C, Zhang Y, Wang S, Jiang S, Zheng J, Yin C, Zhang C. Regulator of G-protein signaling 14 (RGS14) promotes cancer growth in hepatocellular carcinoma. Cancer Genet 2025; 294-295:80-89. [PMID: 40245482 DOI: 10.1016/j.cancergen.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major contributor to cancer-related deaths globally. The progression of HCC is influenced by a range of intrinsic and extrinsic factors, necessitating further research into the molecular mechanisms involved. While Regulator of G-protein Signaling 14 (RGS14) has shown emerging roles in cancer biology, its function in HCC remains poorly characterized. MATERIALS AND METHODS RGS14 expression and clinical significance were analyzed using TCGA-LIHC, HCCDB, and GEO datasets. Immunofluorescence (IF) staining was employed to validate protein expression. Functional assays, including cell proliferation, migration, invasion, and in vivo xenograft models, were conducted to assess the oncogenic role of RGS14. Bulk-mRNA sequencing was performed using in situ tumor tissues to identify RGS14-regulated pathways. RESULTS RGS14 was significantly upregulated in HCC tissues and positively associated with poor patient outcomes. In vitro experiments demonstrated that RGS14 enhanced HCC cell proliferation, migration, and invasion, while in vivo studies confirmed its tumor-promoting effects. Mechanistically, RGS14 activated the extracellular matrix (ECM)-receptor interaction pathway to drive HCC progression. CONCLUSION Our findings suggest that RGS14 could serve as a novel prognostic marker and therapeutic target for HCC, contributing to improved treatment strategies.
Collapse
Affiliation(s)
- Yi Ran
- Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China; Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Liping Li
- Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Zhihua Wang
- Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Ting Sun
- Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Cong Wen
- Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Yixin Zhang
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Shu Wang
- Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Shishi Jiang
- Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Junjie Zheng
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Changjun Yin
- Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Chuankai Zhang
- Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China; Precision Medicine Institute, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, PR China.
| |
Collapse
|
21
|
Gao X, Xu Y, Hu X, Chen J, Zhang D, Xu X. Comprehensive analysis of mitochondrial solute carrier family 25 (SLC25) identifies member 19 (SLC25A19) as a regulatory factor in hepatocellular carcinoma. Gene 2025; 944:149299. [PMID: 39892835 DOI: 10.1016/j.gene.2025.149299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/08/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The mitochondrial solute carrier family 25 (SLC25) is known to play a pivotal role in oncogenesis, yet its specific involvement in hepatocellular carcinoma (HCC) remains poorly elucidated. METHODS In this study, we performed a clustering analysis of HCC patients in the Cancer Genome Atlas database based on the expression levels of SLC25 members, and conducted clinical feature analysis for each patient within the clusters. Subsequently, we developed a prognostic model using a Lasso regression approach with SLC25A19, SLC25A49, and SLC25A51 as features, and generated a risk score for each HCC patient. We then identified SLC25A19 as a potential prognostic marker for HCC through single-cell analysis, and validated this finding using in vitro and in vivo experiments. RESULTS Our results revealed significant differences in the expression of most SLC25 family members in HCC patients, enabling the stratification of patients into three clusters, with those in cluster 1 exhibiting the most favorable prognosis and showing a correlation with enhanced immune infiltration. The risk scores derived from the features SLC25A19, SLC25A49, and SLC25A51 effectively predicted the prognosis of HCC patients, with area under the curve (AUC) values exceeding 0.7 in the test group. Single-cell analysis further demonstrated h eightened expression of SLC25A19 in the immune microenvironment of HCC, and in vitro experiments indicated that SLC25A19 may regulate the proliferation, migration, invasion, cycle, and apoptosis of liver cancer cells through the Wnt pathway. In the HepG2 animal model, overexpression of SLC25A19 significantly promotes tumor growth, while knockdown inhibits tumor growth. Analysis of patient tumor tissues shows that SLC25A19 is highly expressed in liver cancer tissues and is associated with CD8+ T cell infiltration. CONCLUSIONS In conclusion, our comprehensive analysis of the role of SLC25 in HCC unveiled SLC25A19 as a potential regulatory factor in HCC.
Collapse
Affiliation(s)
- Xueke Gao
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Yangtao Xu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Xinyao Hu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Jiayu Chen
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Daoming Zhang
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Ximing Xu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060.
| |
Collapse
|
22
|
Zhao Y, Wang X, Yang X, Li J, Han B. Insights into the history and trends of nanotechnology for the treatment of hepatocellular carcinoma: a bibliometric-based visual analysis. Discov Oncol 2025; 16:484. [PMID: 40192866 PMCID: PMC11977073 DOI: 10.1007/s12672-025-02145-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/13/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Nanotechnology has great potential and advantages in the treatment of hepatocellular carcinoma (HCC), but the research trends and future directions are not yet clear. OBJECTIVES Analyze the development trajectory, research hotspots, and future trends of nanotechnology and HCC research globally in the past 20 years, providing a more comprehensive and intuitive reference for researchers in this field. METHODS Retrieve relevant literature on nanotechnology and HCC research in the Web of Science (WOS) Core Collection database, and conduct bibliometric analysis using software such as CiteSpace, VOSviewer, and SCImago Graphica. RESULTS A total of 852 English publications meeting the criteria were retrieved from the WOS database, with an overall increasing trend in the number of publications and citation frequency over the years. China leads in the number of publications and international collaborations, followed by the USA and India. The most influential research institution is the Chinese Academy of Sciences, the most influential scholar/team is the Rahman, Mahfoozur team, and the journal with the most publications is the International Journal of Nanomedicine. A comprehensive analysis reveals that the current main research directions include new types of nanoparticles, targeted drug delivery systems, photothermal/photodynamic therapy, gene delivery systems, diagnostics, and imaging. It is anticipated that further collaboration among scholars, institutions, and countries will accelerate the development of nanotechnology in the field of HCC research. CONCLUSION This study provides an in-depth analysis of the research status and development trends of nanotechnology in treating HCC from a bibliometric perspective, offering possible guidance for researchers to explore hot topics and frontiers, select suitable journals, and partners in this field.
Collapse
Affiliation(s)
- Yulei Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Xingxin Wang
- College of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaoman Yang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Jiaheng Li
- College of Health, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Bingbing Han
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
23
|
Tang H, Zi H, Zhou D, Li Y, Li X, Chen Z, Zhu Q, Ouyang Q, He P, Chen S, Li Y, Long J, Huang J. Role of the nucleotide excision repair function of CETN2 in the inhibition of the sensitivity of hepatocellular carcinoma cells to oxaliplatin. Carcinogenesis 2025; 46:bgaf003. [PMID: 39945187 DOI: 10.1093/carcin/bgaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/20/2024] [Accepted: 01/18/2025] [Indexed: 04/22/2025] Open
Abstract
Resistance to platinum-based chemotherapy agents like oxaliplatin (OXA) poses significant challenges in the treatment of cancers such as hepatocellular carcinoma (HCC). Centrin 2 (CETN2), which functions in nucleotide excision repair (NER) of DNA damage, is overexpressed in HCC. We investigated the potential role of CETN2 in modulating the sensitivity of HCC cells to OXA. CETN2 expression correlated with decreased OXA sensitivity in Huh7 and Hep3B HCC cell lines. CETN2 forms a complex with XPC, which is crucial for the initial DNA damage recognition in NER, thereby enhancing NER and reducing the efficacy of OXA. siRNA-mediated knockdown of CETN2 increased OXA-induced cytotoxicity and apoptosis, confirming its role in chemoresistance. Moreover, overexpression of CETN2 inhibited OXA-induced DNA damage, an effect partially reversed by XPC knockdown. Our findings highlight CETN2 as a potential biomarker and therapeutic target in overcoming OXA resistance in HCC and suggest the possibility for CETN2 inhibitors in enhancing chemotherapeutic efficacy in the treatment of HCC.
Collapse
Affiliation(s)
- Hengcheng Tang
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Huaduan Zi
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Donghu Zhou
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yanmeng Li
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xiaojin Li
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhibin Chen
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Qianyu Zhu
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Qin Ouyang
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Pingping He
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Sisi Chen
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yanling Li
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jiang Long
- Beijing Minimally Invasive Oncology Medical Center of Traditional Chinese and Western Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101121, China
| | - Jian Huang
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
24
|
Liu Q, Zhang X, Qi J, Tian X, Dovjak E, Zhang J, Du H, Zhang N, Zhao J, Zhang Y, Wang L, Wei Y, Liu C, Qian R, Xiang L, Li W, Xiu P, Ma C, Yu Y, Jiang S. Comprehensive profiling of lipid metabolic reprogramming expands precision medicine for HCC. Hepatology 2025; 81:1164-1180. [PMID: 38899975 PMCID: PMC11902616 DOI: 10.1097/hep.0000000000000962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 05/11/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND AIMS Liver HCC is the second leading cause of cancer-related deaths worldwide. The heterogeneity of this malignancy is driven by a wide range of genetic alterations, leading to a lack of effective therapeutic options. In this study, we conducted a systematic multi-omics characterization of HCC to uncover its metabolic reprogramming signature. APPROACH AND RESULTS Through a comprehensive analysis incorporating transcriptomic, metabolomic, and lipidomic investigations, we identified significant changes in metabolic pathways related to glucose flux, lipid oxidation and degradation, and de novo lipogenesis in HCC. The lipidomic analysis revealed abnormal alterations in glycerol-lipids, phosphatidylcholine, and sphingolipid derivatives. Machine-learning techniques identified a panel of genes associated with lipid metabolism as common biomarkers for HCC across different etiologies. Our findings suggest that targeting phosphatidylcholine with saturated fatty acids and long-chain sphingolipid biosynthesis pathways, particularly by inhibiting lysophosphatidylcholine acyltransferase 1 ( LPCAT1 ) and ceramide synthase 5 ( CERS5 ) as potential therapeutic strategies for HCC in vivo and in vitro. Notably, our data revealed an oncogenic role of CERS5 in promoting tumor progression through lipophagy. CONCLUSIONS In conclusion, our study elucidates the metabolic reprogramming nature of lipid metabolism in HCC, identifies prognostic markers and therapeutic targets, and highlights potential metabolism-related targets for therapeutic intervention in HCC.
Collapse
Affiliation(s)
- Qingbin Liu
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
- Post-Doctoral Research Center, Cisen Pharmaceutical Co. Ltd, Jining, Shandong, China
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Xiangyu Zhang
- Pathology Department, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Jingjing Qi
- Medical Faculty, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Xinchen Tian
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Eva Dovjak
- Medical Faculty, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Jiaqi Zhang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Honghuan Du
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Ni Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jing Zhao
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Yiming Zhang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Lijuan Wang
- Department of Ultrasonic Medicine, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Yangang Wei
- Post-Doctoral Research Center, Cisen Pharmaceutical Co. Ltd, Jining, Shandong, China
| | - Chenqiao Liu
- Hepatobiliary Surgery Department, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Ruikun Qian
- Hepatobiliary Surgery Department, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Longquan Xiang
- Pathology Department, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Weiyang Li
- School of Biological Sciences, Jining Medical University, Rizhao, Shandong, China
| | - Peng Xiu
- Department of General Surgery, Shandong Province Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, Shandong, China
| | - Changlin Ma
- Hepatobiliary Surgery Department, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| | - Yong Yu
- Medical Faculty, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, China
| |
Collapse
|
25
|
Wang Y, Chi S, Tian Y, Li X, Zhang H, Xu Y, Huang C, Gao Y, Jin G, Fu Q, Cao W, Chen C, Ding H, Zhang Y, Hong Y, Li J, Sun X, Li E, Zhang Y, Yao W, Liu R, Hua Y, Huang H, Xu M, Zhang B, Tao W, Yang T, Gao Y, Wang X, Lin C, Li J, Zhang Q, Liang T. Construction of an artificially intelligent model for accurate detection of HCC by integrating clinical, radiological, and peripheral immunological features. Int J Surg 2025; 111:2942-2952. [PMID: 39878177 DOI: 10.1097/js9.0000000000002281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Integrating comprehensive information on hepatocellular carcinoma (HCC) is essential to improve its early detection. We aimed to develop a model with multimodal features (MMF) using artificial intelligence (AI) approaches to enhance the performance of HCC detection. MATERIALS AND METHODS A total of 1092 participants were enrolled from 16 centers. These participants were allocated into the training, internal validation, and external validation cohorts. Peripheral blood specimens were collected prospectively and subjected to mass cytometry analysis. Clinical and radiological data were obtained from electrical medical records. Various AI methods were employed to identify pertinent features and construct single-modal models with optimal performance. The XGBoost algorithm was utilized to amalgamate these models, integrating multimodal information and facilitating the development of a fusion model. Model evaluation and interpretability were demonstrated using the SHapley Additive exPlanations method. RESULTS We constructed the electronic health record, BioScore, RadiomicScore, and DLScore models based on clinical, radiological, and peripheral immunological features, respectively. Subsequently, these single-modal models were amalgamated to develop an all-in-one MMF model. The MMF model exhibited enhanced performance compared to models comprising only single-modal features in detecting HCC. This superiority in performance was confirmed through the internal and external validation cohorts, yielding area under the curve (AUC) values of 0.985 and 0.915, respectively. Additionally, the MMF model improved the detection ability in subpopulations of HCCs that were negative for alpha-fetoprotein and those with small size, with AUC values of 0.974 and 0.996, respectively. CONCLUSIONS Integrating MMF improved the performance of the model for HCC detection.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengqiang Chi
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yu Tian
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xueyao Li
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Hang Zhang
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Yiting Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Huang
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Yiwei Gao
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Gaowei Jin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qihan Fu
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wanyue Cao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cao Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haonan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yupeng Hong
- Department of Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Junjian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xu Sun
- Department of General Surgery, Huzhou Central Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Enliang Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuhua Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
| | - Weiyun Yao
- Department of Surgery, Changxing People's Hospital, Huzhou, China
| | - Runtian Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongfei Hua
- Department of General Surgery, Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, China
| | - Haifeng Huang
- Department of General Surgery, Shengzhou People's Hospital, Shengzhou, China
| | - Minghui Xu
- Department of General Surgery, Haining People's Hospital, Haining, China
| | - Bo Zhang
- Department of General Surgery, Shenzhen University Luohu People's Hospital, Shenzhen, China
| | - Weifeng Tao
- Department of General Surgery, Shangyu People's Hospital of Shaoxing, Shangyu, China
| | - Tianxing Yang
- Department of Medical Oncology, Sanmen People's Hospital, Taizhou, China
| | - Yuming Gao
- Department of General Surgery, Jixi County People's Hospital, Jixi, China
| | - Xiaoguang Wang
- Department of General Surgery, Jiaxing Second People's Hospital, Jiaxing, China
| | - Cheng Lin
- Zhejiang Puluoting Health Technology Co Ltd, Hangzhou, China
| | - Jingsong Li
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Cancer Center of Zhejiang University, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Cancer Center of Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Alsaqa M, Sierra L, Marenco-Flores A, Parraga X, Barba R, Goyes D, Ozturk NB, Curry MP, Bonder A, Saberi B. Metabolic dysfunction-associated steatotic liver disease correlates with higher lower graft survival in liver transplant recipients with hepatocellular carcinoma. Eur J Gastroenterol Hepatol 2025; 37:497-504. [PMID: 39976068 DOI: 10.1097/meg.0000000000002914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BACKGROUND Direct-acting antivirals (DAAs) have revolutionized hepatitis C virus (HCV) treatment. The changing landscape of hepatocellular carcinoma (HCC) in liver transplant (LT) recipients lacks a thorough description of the outcomes of HCC based on etiology. OBJECTIVE To assess the waitlist (WL) dropout and graft survival in HCC LT candidates based on the etiology of HCC in the post-DAA era. METHODS This retrospective cohort study analyzed United Network Organ Sharing/Organ Procurement Transplant Network data from 2015 to 2022. Graft survival was analyzed using Kaplan-Meier curves, and predictors of WL dropout and graft failure were assessed using multivariate analysis. RESULTS Among LT recipients, etiologies were HCV (53.6%), alcohol-associated liver disease (ALD) (12.0%), metabolic dysfunction-associated steatotic liver disease (MASLD) (16.6%), hepatitis B virus (HBV) (5.6%), and other (12.1%). MASLD and ALD had the highest dropout rates (1-year: 20.4%, 21.7%; 3-year: 58.2%, 51.1%; P < 0.001). Dropout was linked to diabetes, low albumin, high Model of End-Stage Liver Disease, high alpha-fetoprotein, tumor number, and size. MASLD had the worst 1-, 3-, and 5-year graft survival (89.8%, 81.8%, and 74.1%) and higher failure risk than HCV (hazard ratio: 1.143, 95% CI: 1.021-1.281). Diabetes negated MASLD's impact on graft failure but worsened survival for MASLD-HCC compared with HBV and ALD, matching HCV. CONCLUSION MASLD has the highest WL dropout and post-LT graft failure among HCC LT candidates, surpassing HCV in the post-DAA era. The worst graft survival in MASLD-HCC is associated with pre-LT diabetes.
Collapse
Affiliation(s)
- Marwan Alsaqa
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Leandro Sierra
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ana Marenco-Flores
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ximena Parraga
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Romelia Barba
- Department of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Daniela Goyes
- Division of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut
| | - N Begum Ozturk
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, Michigan, USA
| | - Michael P Curry
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Alan Bonder
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Behnam Saberi
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Huang C, Li Y, Zhang F, Zhang C, Ding Z. Advancements in elucidating the mechanisms of Sorafenib resistance in hepatocellular carcinoma. Int J Surg 2025; 111:2990-3005. [PMID: 39992113 DOI: 10.1097/js9.0000000000002294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025]
Abstract
Primary liver cancer is a major global health challenge, of which hepatocellular carcinoma is the most common. For patients with advanced liver cancer, Sorafenib is a first-line targeted drug that occupies a dominant position in clinical applications. Sorafenib is a multi-kinase inhibitor commonly used in clinical practice, which can effectively inhibit tumor cell proliferation, promote cell apoptosis, and inhibit angiogenesis. However, the emergence of drug resistance has hindered the development of treatment programs, which is an urgent problem to be solved. Recent studies have revealed many mechanisms and influencing factors of Sorafenib resistance (such as epigenetic regulation, programmed cell death, metabolic reprogramming, and tumor microenvironment changes). This review not only summarizes the above mechanisms, but also summarizes the combined application of Sorafenib with other drugs (such as molecular targeted drugs, other anti-angiogenesis drugs, cytotoxic drugs, immunotherapy drugs, etc .). Finally, potential strategies and research directions to overcome drug resistance (such as targeting epigenetic pathways or metabolic reprogramming) are discussed to provide suggestions for future in-depth research and clinical applications.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangqian Li
- Frontiers Science Center for Disease-related Molecular Network, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fengmei Zhang
- Frontiers Science Center for Disease-related Molecular Network, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenliang Zhang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenyu Ding
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
28
|
Wang C, Liu J, Wu Y, Cai C, Chai Z, Jia P, Yuan Y, Jiang Z. AURKB as a Therapeutic Target and Key Driver of Liver Cancer Growth and Metastasis. APMIS 2025; 133:e70021. [PMID: 40177797 DOI: 10.1111/apm.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/05/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths worldwide. Aurora kinase B (AURKB), a critical regulator of mitosis, has been implicated in cancer progression, though its precise role in HCC remains unclear. In this study, AURKB expression was found to be significantly elevated in HCC tissues and cell lines compared to controls, as validated by GEPIA and ENCORI databases. Functional assays revealed that AURKB knockdown reduced cell proliferation, invasion, and migration, while increasing apoptosis. Furthermore, suppression of AURKB affected epithelial-mesenchymal transition (EMT) markers, decreasing vimentin and N-cadherin levels and increasing E-cadherin expression. In vivo, a xenograft mouse model demonstrated that tumors derived from AURKB-silenced cells exhibited reduced growth and fewer lung metastases. Histological and immunohistochemical analyses showed lower levels of Ki-67, MMP-9, and EMT markers in these tumors, alongside increased E-cadherin. These findings highlight AURKB's critical role in promoting HCC progression, metastasis, and EMT regulation. Overexpression of AURKB was associated with poor prognosis, suggesting it could serve as a potential biomarker and therapeutic target for liver cancer. Overall, targeting AURKB may provide a novel approach to inhibit HCC growth and metastasis, improving patient outcomes.
Collapse
Affiliation(s)
- Chen Wang
- Department of Interventional Vascular, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Jiangwen Liu
- Department of Hepatobilary Surgery, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Yali Wu
- Department of Insurance Office, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Chen Cai
- Department of Interventional Vascular, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Zhiwei Chai
- Department of Medical, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Ping Jia
- Department of Catheterization Room, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Yueyue Yuan
- Department of Interventional Vascular, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| | - Zhixin Jiang
- Department of Hepatobilary Surgery, The Third Affiliated Hospital of Xinjiang Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
29
|
Ito T, Shimose S, Tani J, Tomonari T, Saeki I, Takeuchi Y, Hatanaka T, Sasaki K, Kakizaki S, Kanayama Y, Yoshioka N, Naito T, Takeuchi M, Yasunaka T, Sakata M, Iwamoto H, Itano S, Shirono T, Tanabe N, Yamamoto T, Naganuma A, Nishina S, Otsuka M, Takami T, Takayama T, Kawaguchi T, Kawashima H. Antitumor effects and immune-mediated adverse events of durvalumab plus tremelimumab treatment for unresectable hepatocellular carcinoma. Hepatol Res 2025; 55:577-587. [PMID: 40318087 DOI: 10.1111/hepr.14151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
AIM Durvalumab plus tremelimumab (Dur/Tre) is a first-line systemic treatment option for unresectable hepatocellular carcinoma (uHCC). However, the management of severe immune-mediated adverse events (imAEs) is challenging. Therefore, we investigated the relationship between severe imAEs and antitumor responses in patients with uHCC treated with Dur/Tre. METHODS We included 157 patients with uHCC treated with Dur/Tre in this multicenter, retrospective study and analyzed the relationship between progression-free survival (PFS)/antitumor response and severe imAEs requiring high-dose corticosteroid treatment. RESULTS Thirty-two patients (20.4%) developed severe imAEs, including enterocolitis/diarrhea (n = 10), liver injury (n = 9), interstitial lung disease (n = 5), rashes (n = 4), cytokine-release syndrome/fever (n = 2), pancreatitis (n = 2), and others (n = 4) (median follow-up period, 6.8 months). Infliximab was administered in six patients with steroid-refractory enterocolitis. Although the objective response rate (ORR) and disease control rate (DCR) were significantly higher with first-line therapy than with later-line therapy (p = 0.026), the frequency of severe imAEs was not significantly different (p = 0.221). The ORR and DCR in patients with and without severe imAEs were 15.6% and 17.6% and 65.6% and 47.2%, respectively, with no significant differences. Five patients with severe imAEs, including rashes and liver injury, showed objective responses (partial response + complete response). Among patients who achieved an objective response, the PFS at 10 months was good (100% and 70.3% with and without high-dose corticosteroids, respectively). CONCLUSIONS Severe imAEs of Dur/Tre treatment requiring high-dose corticosteroid treatment did not affect antitumor efficacy, which differed depending on the type of imAEs. Therefore, appropriately managing imAEs is essential to guide sequential treatment.
Collapse
Affiliation(s)
- Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Nagoya, Japan
| | - Shigeo Shimose
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Joji Tani
- Faculty of Medicine, Department of Gastroenterology and Neurology, Kagawa University, Takamatsu, Kagawa, Japan
| | - Tetsu Tomonari
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Issei Saeki
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yasuto Takeuchi
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Kyo Sasaki
- Department of Gastroenterology and Hepatology, Kawasaki Medical School, Kurashiki, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Yuki Kanayama
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Naoki Yoshioka
- Department of Gastroenterology and Hepatology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Japan
| | - Takehito Naito
- Department of Gastroenterology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Mamiko Takeuchi
- Department of Gastroenterology, Anjo Kosei Hospital, Anjo, Japan
| | - Tetsuya Yasunaka
- Department of Gastroenterology, Fukuyama City Hospital, Fukuyama, Japan
| | - Masahiro Sakata
- Department of Gastroenterology, National Hospital Organization Fukuyama Medical Center, Fukuyama, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Department of Gastroenterology and Hepatology, Iwamoto Internal Medical Clinic, Kitakyusyu, Japan
| | - Satoshi Itano
- Department of Gastroenterology and Hepatology, Kurume Central Hospital, Kurume, Japan
| | - Tomotake Shirono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Norikazu Tanabe
- Division of Laboratory, Yamaguchi University Hospital, Ube, Japan
| | - Takafumi Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Nagoya, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Sohji Nishina
- Department of Gastroenterology and Hepatology, Kawasaki Medical School, Kurashiki, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Taro Takami
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
30
|
Jain NK, Chandrasekaran B, Khazaleh N, Jain HK, Lal M, Joshi G, Jha V. Computational Network Pharmacology, Molecular Docking, and Molecular Dynamics to Decipher Natural Compounds of Alchornea laxiflora for Liver Cancer Chemotherapy. Pharmaceuticals (Basel) 2025; 18:508. [PMID: 40283942 PMCID: PMC12030508 DOI: 10.3390/ph18040508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Background:Alchornea laxiflora (Benth.) Pax & K. Hoffm. (A. laxiflora) is utilized as a traditional herb for treating several diseases. Objective: Our study aims to identify the active phytochemical candidates from A. laxiflora and analyses to predict their anticancer activity mechanism by employing network pharmacology, molecular docking, and molecular dynamics (MD). Methods: The phytoconstituents of A. laxiflora were retrieved from the literature, and phytoconstituent-related targets implicated in hepatocellular carcinoma (HCC) were collected from respective databases. Computational methods were employed to recognize essential compounds, hub gene targets, and signaling pathways. Results: Our study has identified 12 potentially bioactive compounds, 150 potential anti-HCC targets, and 15 hub gene targets for A. laxiflora. Molecular docking results recognized the better binding energy values of below -5.6 kcal/mol. Further, MD simulations of the three of the top-scoring protein-ligand complexes (MAPK-3-acetylursolic acid, AKT1-quercetin, and AKT1-3-acetylursolic acid) allowed us to validate the docking results, evaluate the stability of the complexes, and associated conformational changes. Conclusions: Our research claims that phytoconstituents of A. laxiflora are crucial for treating liver cancer, and the recognized protein targets can serve as biomarkers, respectively.
Collapse
Affiliation(s)
- Nem Kumar Jain
- School of Pharmacy, ITM University, Gwalior 474001, Madhya Pradesh, India;
| | | | - Nasha’t Khazaleh
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan;
| | - Hemant Kumar Jain
- Department of General Medicine, Government Medical College, Datia 475661, Madhya Pradesh, India
| | - Moti Lal
- School of Sciences, ITM University, Gwalior 474001, Madhya Pradesh, India;
| | - Gaurav Joshi
- Department of Pharmaceutical Science, Hemvati Nandan Bahuguna Garhwal University, Srinagar 246174, Uttarakhand, India;
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Vibhu Jha
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
31
|
Du D, Zhang W, Zhang D, Liu L, Li J, Chen Z, Yu X, Ye M, Wang W, Li Z, Shao J. NUB1 reduction promotes PCNA-mediated tumor growth by disturbing the PCNA polyubiquitination/NEDDylation in hepatocellular carcinoma cells. Cell Death Dis 2025; 16:228. [PMID: 40164590 PMCID: PMC11958677 DOI: 10.1038/s41419-025-07567-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/08/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Negative regulator of ubiquitin-like protein 1 (NUB1), an inhibitor of neural precursor cells expressed developmentally downregulated 8 (NEDD8), is implicated in tumor growth. However, the expression of NUB1 in hepatocellular carcinoma (HCC) and its effects on HCC growth remain unclear. In this study, our findings revealed reduced NUB1 protein expression in HCC tissues and cells, leading to increased proliferating cell nuclear antigen (PCNA) protein stability through upregulating NEDD8 to promote HCC cell growth. Mechanistically, NUB1 reduction upregulated NEDD8 to promote PCNA NEDDylation at lysine 164 (Lys164), in turn, antagonized PCNA K48-linked polyubiquitination, thereby increasing the stability of PCNA in HCC cells. Finally, the results of the in vitro and in vivo experiments revealed that the NEDDylation inhibitor TAS4464 could inhibit PCNA NEDDylation to decrease PCNA protein expression, thereby suppressing HCC cell growth. Collectively, our results identified NUB1 as a negative regulator of HCC proliferation and confirmed that PCNA NEDDylation promotes PCNA protein stability by antagonizing PCNA polyubiquitination. This study provides a new perspective on the specific mechanism of HCC growth. It expands our understanding of the role of NEDDylation in the regulation of substrate proteins and their functions.
Collapse
Affiliation(s)
- Dongnian Du
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Wenming Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Dandan Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Lingpeng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Jiajuan Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Zehao Chen
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Xuzhe Yu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Miao Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Wei Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Zijing Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Jianghua Shao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China.
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China.
- Liver Cancer Institute, Nanchang University, Nanchang, 330000, China.
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Nanchang University, Nanchang, China.
| |
Collapse
|
32
|
Wang Y, Zhu XF, Gu WJ, Zhang GH. Alterations of the microenvironment of hepatocellular carcinoma in different unfolded protein response activity states. Discov Oncol 2025; 16:393. [PMID: 40133716 PMCID: PMC11937449 DOI: 10.1007/s12672-025-02164-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The unfolded protein response (UPR) is an adaptive and cytoprotective sensing-signaling network. Numerous studies have indicated the crucial role of UPR in the anti-tumor drug resistance and the modification of tumor microenvironment (TME). The aim of this study is to analyze the alterations of microenvironment and key regulatory genes in hepatocellular carcinoma (HCC) with high UPR activity. METHODS We profiled differentially expressed genes (DEGs) by UPR activity, and the biological functions of DEGs and the alterations of signaling pathways were explored. The Immune/Stromal scores and relative abundance of infiltrating cells of HCC tissues with RNA sequencing data downloaded from The Cancer Genome Atlas (TCGA) were calculated by the xCell and ESTIMATE algorithm. The correlations between the prognostic UPR-related genes with the microenvironment scores and infiltrating cells were analyzed using R package "corrplot". RESULTS Our results demonstrated that UPR-related genes mainly involved in immune-related signaling pathways. Microenvironment analysis revealed that HCC tissues with higher UPR activity had lower Stromal scores and the relative abundance of various infiltrating cells including hematopoietic stem cells (HSC), lymphatic endothelial cells (LECs), microvascular endothelial cells, endothelial cells (ECs) and adipocytes decreased most significantly. Kaplan-Meier survival analysis indicated that the decline of Stromal scores and corresponding infiltrating stromal cells would result in worse prognosis. The expression levels of CLEC3B, RAMP3, GPR182 and DNASE1L3 were significantly positively correlated with Stromal scores and various infiltrating stromal cells, and down-regulation of these genes were also associated with worse prognosis of HCC. CONCLUSIONS HCC with high UPR activity had lower Stromal scores and worse prognosis. Down-regulated genes CLEC3B, RAMP3, GPR182 and DNASE1L3 may play an important regulatory role in the modification of microenvironment of HCC with high UPR activity.
Collapse
Affiliation(s)
- Yao Wang
- Department of Clinical Laboratory, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, No. 155, Han'Zhong Road, Qinhuai, Nanjing, 210029, Jiangsu, China
| | - Xiao Fei Zhu
- Department of Clinical Laboratory, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, No. 155, Han'Zhong Road, Qinhuai, Nanjing, 210029, Jiangsu, China
| | - Wan Jian Gu
- Department of Clinical Laboratory, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, No. 155, Han'Zhong Road, Qinhuai, Nanjing, 210029, Jiangsu, China
| | - Gui Hong Zhang
- Department of Clinical Laboratory, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, No. 155, Han'Zhong Road, Qinhuai, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
33
|
Cannet F, Sequera C, Veloso PM, El Kaoutari A, Methia M, Richelme S, Kaya M, Cherni A, Dupont M, Borg JP, Morel C, Boursier Y, Maina F. Tracing specificity of immune landscape remodeling associated with distinct anticancer treatments. iScience 2025; 28:112071. [PMID: 40124507 PMCID: PMC11930375 DOI: 10.1016/j.isci.2025.112071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/18/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Immune cells within the tumor microenvironment impact cancer progression, resistance, response to treatments. Despite remarkable outcomes for some cancer patients, immunotherapies remain unsatisfactory for others. Here, we designed an experimental setting using the Alb-R26 Met "inside-out" mouse model, faithfully recapitulating molecular features of liver cancer patients, to explore the effects of distinct anticancer targeted therapies on the tumor immune landscape. Using two treatments in clinical trials for different cancer types, Decitabine and MEK+BCL-XL blockage, we show their capability to trigger tumor regression in Alb-R26 Met mice and to superimpose distinct profiles of immune cell types and immune-checkpoints, impacting immunotherapy response. A machine learning approach processing tumor imaging and immune profile data identified a putative signature predicting tumor treatment response in mice and patients. Outcomes exemplify how the tumor immune microenvironment is differentially reshaped by distinct anticancer agents and highlight the importance of measuring its modulation during treatment to optimize oncotherapy and immunotherapy combinations.
Collapse
Affiliation(s)
- Floriane Cannet
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Célia Sequera
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Paula Michea Veloso
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Abdessamad El Kaoutari
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Melissa Methia
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Sylvie Richelme
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Muge Kaya
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Afef Cherni
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Mathieu Dupont
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Jean-Paul Borg
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Institut Universitaire de France, Paris, France
| | - Christian Morel
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | | | - Flavio Maina
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| |
Collapse
|
34
|
Wang J, Wang X, Shi J, Wang Y, Mi L, Zhao M, Han G, Yin F. CD161, a promising prognostic biomarker in hepatocellular carcinoma, correlates with immune infiltration. PeerJ 2025; 13:e19055. [PMID: 40115278 PMCID: PMC11925045 DOI: 10.7717/peerj.19055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/05/2025] [Indexed: 03/23/2025] Open
Abstract
Background CD161, encoded by the killer cell lectin-like receptor B1 (KLRB1) gene, exhibits varied roles among different tumors. This study aimed to explore both the potential value of CD161 as a prognostic biomarker for hepatocellular carcinoma (HCC) and its association with immune cell infiltration. Methods A total of 109 HCC patients who underwent surgery were retrospectively analyzed. Immunohistochemistry, bioinformatic analyses, and statistical measurements were used to investigate the associations between CD161 expression, immune cell infiltration, and clinical outcomes in both public databases and in-house cohorts. Results CD161 was highly expressed at both protein and mRNA levels in adjacent normal tissues compared to tumor tissues of HCC patients. Meanwhile, CD161 was enriched in HCC cases characterized by smaller tumor sizes (≤5 cm) and the absence of portal vein tumor thrombus. Individuals with high CD161 expression showed extended overall survival (OS) and relapse free survival (RFS) compared to those with lower CD161 levels. CD161 was identified as an independent prognostic indicator for both OS and RFS. In addition, the enrichment analysis indicated a close correlation between CD161 and immune response, as well as between CD161 and the signaling pathways of cytokines and chemokines, implying its role in immune regulation during cancer development. Specifically, CD161 expression was positively associated with immunomodulators and tumor-infiltrating immune cells, especially CD8+T cells, CD4+T cells, and dendritic cells. Multiple public databases showed that patients with high CD161 expression were more likely to derive benefits from immunotherapy. Conclusion CD161 was identified as a promising prognostic biomarker for HCC, as its expression indicates a favorable prognosis. Additionally, CD161 is closely linked to high infiltration of immune cells, participates in the regulation of the tumor immune microenvironment, and holds promise as a potential biomarker for predicting the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Jinfeng Wang
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoxiao Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianfei Shi
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongjun Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lili Mi
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Man Zhao
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guangjie Han
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fei Yin
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
35
|
Mui S, Shi J, Wen K, Yan Y, Li H, Wang W, Zhou Z, Xiao Z. Multi-omics analysis identifies OSGEPL1 as an oncogene in hepatocellular carcinoma. Discov Oncol 2025; 16:328. [PMID: 40090949 PMCID: PMC11911280 DOI: 10.1007/s12672-025-02066-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
PURPOSE N6-Threonylcarbamoyladenosine (t6A) modification irregularities and their associated enzymes genes (OSGEP, OSGEPL1, TPRKB, GON7, TP53RK, YRDC, and LAGE3) are linked to various malignancies development, including Hepatocellular Carcinoma (HCC), yet the specific mechanisms remain obscure. This gap in knowledge is significant, as understanding the mechanisms of t6A modification could reveal new insights into HCC pathogenesis and potentially identify novel therapeutic targets. METHODS We leveraged data from The Cancer Genome Atlas (TCGA) to analyze the expression of t6A-associated genes, with a focus on OSGEPL1 in HCC. Our analyses included survival outcome, gene expression, functional enrichment, immune cell infiltration, and somatic mutation data. RESULTS We discovered that OSGEPL1 is upregulated in HCC and is correlated with tumor grade, pathological T stage, and overall stage. It inversely impacts overall survival and immune cell infiltration. In vitro experiments confirmed the role of OSGEPL1 in promoting HCC cell proliferation. CONCLUSIONS This study implicates t6A modification pathway dysregulation in HCC prognosis, identifying OSGEPL1 as a potential therapeutic target. These findings provide novel insights into HCC pathogenesis and may guide future treatment strategies.
Collapse
Affiliation(s)
- Sintim Mui
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Juanyi Shi
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Kai Wen
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Yongcong Yan
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Huoming Li
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Weidong Wang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Zhenyu Zhou
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
| | - Zhiyu Xiao
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
36
|
Choi SK, Kim MJ, You JS. αKG-induced oxidative stress and mTOR inhibition as a therapeutic strategy for liver cancer. Med Oncol 2025; 42:105. [PMID: 40080333 PMCID: PMC11906577 DOI: 10.1007/s12032-025-02653-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
Despite the availability of targeted therapies, liver cancer remains a severe health burden. The need for adjuvant therapy to improve treatment efficacy and prevent recurrence is emerging. Alpha-ketoglutarate (αKG) is an intermediate in the tricarboxylic acid cycle and a cofactor for various oxygenases. A critical role of this multifunctional metabolite has started to be revealed in physiological and pathological conditions. We found that αKG exerts various anti-tumor effects in liver cancer cells. Our kinetic transcriptome study suggested that increasing reactive oxygen species and inhibiting mTORC1 signaling underlies. Indeed, αKG treatment elevated oxidative stress and induced DNA damage, presumably caused by early downregulation of the antioxidant gene SLC7A11. Further, we validated impaired mTOR signaling and decreased cellular energy production. This unique mechanism underscores αKG's potential as a liver cancer therapy by harnessing oxidative stress and disrupting metabolic signaling. These findings could provide valuable insights into further exploration of αKG as a promising therapeutic agent in liver cancer.
Collapse
Affiliation(s)
- Sung Kyung Choi
- Department of Biochemistry, School of Medicine, Konkuk University, Chungju-Si, 27478, Chungcheongbuk-Do, Korea
| | - Myoung Jun Kim
- Department of Biochemistry, School of Medicine, Konkuk University, Chungju-Si, 27478, Chungcheongbuk-Do, Korea
| | - Jueng Soo You
- Department of Biochemistry, School of Medicine, Konkuk University, Chungju-Si, 27478, Chungcheongbuk-Do, Korea.
- Research Institute of Medical Science, KU Open Innovation Center, Chungju-Si, 27478, Chungcheongbuk-Do, Korea.
| |
Collapse
|
37
|
Ekpenyong BB, Ubi GM, Kooffreh ME, Umoyen AJ, James CS, Ettah IA, Etangetuk NA, Effiom BE, Okpechi PA, Ejue BP, Ambo OA. Tumor protein 53 gene polymorphism, demographic attributes and associated risk factors among liver cancer patients in Calabar, Nigeria. BMC Cancer 2025; 25:430. [PMID: 40065269 PMCID: PMC11892161 DOI: 10.1186/s12885-025-13803-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Mutations in the TP53 gene had been attributed to the development of liver cancer. Hepatocellular carcinoma (HCC) and liver tumour are liver diseases having high mortality rates in several populations. There is no information on the TP53 gene polymorphism among liver diseases patients in Calabar, Nigeria. This study investigated the genetic polymorphism of TP53 among HCC and liver tumour in Calabar. This research was carried out in the University of Calabar Teaching Hospital, Calabar. Blood samples were collected from 35 clinically diagnosed hepatocellular carcinoma and 10 tumour patients and 10 healthy controls. DNA was extracted from all blood samples and Polymerase Chain Reaction (PCR) was performed using specific primers. The PCR amplicons were digested using Hae III restriction enzyme and the genotypic and allelic frequencies was determined. Demographic data among participants showed that males were 68.9% (31), females (31.1%; 14), sex ratio (2.2: 0.5), mean age was 41.51 ± 2.13 years with an odds ratio of 1.25. The distribution of participants according to marital status were 33(73.3%), 10(22.2%), and 2(4.4%) for married, single, and widowed respectively. The participants were from different extractions with varied representations of Yakurr (22.2%, 10), Efik (20%, 9), Boki (13.3%, 6), Ogoja (13.3%, 6), Annang (8.8%, 4), Ibibio (2.2%, 1) and Igbo (2.2%, 1) and respectively. Approximately, 64.7% (30) of the chronic liver diseases were from the Central and Northern part of Cross River State. The risk factors were HCV infection, HBsAg+, alcoholism, smoking, consumption of groundnuts that may have been contaminated with aflatoxin and family history of the disease. PCR product yielded 254 bp and digested PCR product showed homozygous TT mutation (27), heterozygous GT mutation (17) and homozygous GG wild type (1) in cases. The overall TP53 gene mutation frequency was 46.32% (44). The frequency of G allele, T allele, GG, GT and TT were 0.21, 0.79, 0.04, 0.33 and 0.62 respectively among cases, while GG (wild type) was only detected among controls in the study population. The genotypic and allelic frequencies conform to Hardy-Weinberg equilibrium meaning that the forces of evolution were not acting on the locus. There were significant differences in the genotypic proportions of the TP53 gene polymorphism among patients and controls. This study on the TP53 gene polymorphism will serve as baseline information on the molecular etiology of hepatocellular carcinoma and liver tumour in Cross River State.
Collapse
Affiliation(s)
- Blessing B Ekpenyong
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
- Department of Plant Science and Biotechnology, University of Cross River State, Calabar, Nigeria
| | - Godwin M Ubi
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria.
| | - M E Kooffreh
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Anthony J Umoyen
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Cecilia S James
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Ivon A Ettah
- Department of Science Laboratory Technology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Nseabasi A Etangetuk
- Department of Science Laboratory Technology, Faculty of Biological Sciences, University of Calabar, Calabar, Nigeria
| | - Bassey E Effiom
- Department of Guidance and Counseling, Faculty of Education, University of Calabar, Calabar, Nigeria
| | - Philip A Okpechi
- Department of Guidance and Counseling, Faculty of Education, University of Calabar, Calabar, Nigeria
| | - Bassey P Ejue
- Department of Guidance and Counseling, Faculty of Education, University of Calabar, Calabar, Nigeria
| | - Ogar A Ambo
- Department of Guidance and Counseling, Faculty of Education, University of Calabar, Calabar, Nigeria
| |
Collapse
|
38
|
Jeng LB, Chan WL, Teng CF. Independent prognostic significance of tissue and circulating microrna biomarkers in hepatocellular carcinoma. Discov Oncol 2025; 16:281. [PMID: 40056315 DOI: 10.1007/s12672-025-02043-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/04/2025] [Indexed: 03/10/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. Although many therapeutic modalities have been established for treating HCC patients, the outcomes of patients remain unsatisfactory. Development of independent prognostic biomarkers is thus an important need to allow for early diagnosis and timely treatment. MicroRNAs (miRNAs) are the most studied class of small non-coding RNAs. It has been shown that miRNAs play essential roles in the multiple steps of HCC tumorigenesis and progression. Furthermore, the baseline expression levels of many miRNAs are altered in tumor tissues and blood circulation of HCC patients. Therefore, miRNAs have emerged as independent biomarkers for the prediction of HCC prognosis. This review provides a comprehensive literature-based summary of tissue and circulating miRNA biomarkers with independent prognostic significance in HCC.
Collapse
Affiliation(s)
- Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung, 404, Taiwan
- Department of Surgery, China Medical University Hospital, Taichung, 404, Taiwan
- Cell Therapy Center, China Medical University Hospital, Taichung, 404, Taiwan
| | - Wen-Ling Chan
- Department of Public Health, College of Public Health, China Medical University, Taichung, 404, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, 413, Taiwan
| | - Chiao-Fang Teng
- Organ Transplantation Center, China Medical University Hospital, Taichung, 404, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Hsueh-Shih Rd., Northern Dist., Taichung, 404, Taiwan.
- Master Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
39
|
Nosaka T, Naito T, Akazawa Y, Takahashi K, Matsuda H, Ohtani M, Nishizawa T, Okamoto H, Nakamoto Y. Identification of novel antiviral host factors by functional gene expression analysis using in vitro HBV infection assay systems. PLoS One 2025; 20:e0314581. [PMID: 40048440 PMCID: PMC11884705 DOI: 10.1371/journal.pone.0314581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/12/2024] [Indexed: 03/09/2025] Open
Abstract
To cure hepatitis B virus (HBV) infection, it is essential to elucidate the function of hepatocyte host factors in regulating the viral life cycle. Signaling and transcription activator of transcription (STAT)1 play important roles in immune responses, but STAT1-independent pathways have also been shown to have important biological reactivity. Using an in vitro HBV infection assay system, the current study aimed to investigate the STAT1-independent host factors that contribute to the control of viral infection by comprehensive functional screening. The in vitro HBV infection system was established using primary human hepatocytes (PXB cells) infected with HBV derived from a plasmid containing the 1.3-mer HBV genome. Comprehensive functional studies were performed using small interfering RNA (siRNA) and vector transfection and analyzed using microarrays. Knockdown of STAT1 increased viral products in HBV-transfected HepG2 cells, but decreased in HBV-infected PXB cells. RNA microarray was performed using HBV-infected PXB cells with STAT1 knockdown. Fumarylacetoacetate hydrolase (FAH) was extracted by siRNA of genes in PXB cells altered by STAT1 knockdown. Transfection of FAH inhibited HBV replication. Dimethyl fumarate (DMF), the methyl ester of FAH metabolite, showed antiviral effects by inducing autophagy and anti-HBV-related genes. Independently of STAT1, FAH was identified as a host factor that contributes to the control of viral infection, and its metabolite, DMF, exhibited antiviral activity. These results suggest that the novel host factor FAH and its metabolites may be an innovative therapeutic strategy to control the HBV life cycle.
Collapse
Affiliation(s)
- Takuto Nosaka
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tatsushi Naito
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yu Akazawa
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kazuto Takahashi
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hidetaka Matsuda
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masahiro Ohtani
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tsutomu Nishizawa
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Hiroaki Okamoto
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
40
|
Suoangbaji T, Long R, Ng IOL, Mak LLY, Ho DWH. LiverSCA 2.0: An Enhanced Comprehensive Cell Atlas for Human Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. Cancers (Basel) 2025; 17:890. [PMID: 40075736 PMCID: PMC11898674 DOI: 10.3390/cancers17050890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) are two distinct types of primary liver cancer (PLC) characterized by considerable extents of cellular and molecular heterogeneities. We recently developed a web-based cell atlas called LiverSCA that possesses a user-friendly interface and comprehensive functionalities. It facilitates the exploration of gene expression patterns, cellular compositions, and intercellular communication within the microenvironments of liver and PLC tumors. METHODS To further enhance the documentation of data pinpointing different phenotypes/subtypes of liver and PLC, we extended the catalog of LiverSCA with additional datasets, e.g., ICC and metabolic dysfunction-associated steatotic liver disease/steatosis (MASLD/MASH). RESULTS The current enhanced version of the LiverSCA cell atlas encompasses six phenotypes (normal, HBV-HCC, HCV-HCC, non-viral HCC, ICC, and MASH), 63 patients, and over 248,000 cells. Furthermore, we have incorporated comparative visualization methods that allow users to simultaneously examine and compare gene expression levels between two different phenotypes. CONCLUSIONS We are committed to the continuous development of LiverSCA and envision that it will serve as a valuable resource to support researchers in convenient investigations into the cellular and molecular landscapes of liver and PLC.
Collapse
Affiliation(s)
- Tina Suoangbaji
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong; (T.S.); (R.L.); (I.O.-L.N.)
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Hong Kong
| | - Renwen Long
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong; (T.S.); (R.L.); (I.O.-L.N.)
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Hong Kong
| | - Irene Oi-Lin Ng
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong; (T.S.); (R.L.); (I.O.-L.N.)
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Hong Kong
| | - Loey Lung-Yi Mak
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong; (T.S.); (R.L.); (I.O.-L.N.)
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong
| | - Daniel Wai-Hung Ho
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong; (T.S.); (R.L.); (I.O.-L.N.)
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
41
|
Mysko C, Landi S, Purssell H, Allen AJ, Prince M, Lindsay G, Rodrigues S, Irvine J, Street O, Gahloth D, MacLennan S, Piper Hanley K, Hanley N, Athwal VS. Health inequalities in hepatocellular carcinoma surveillance, diagnosis, treatment, and survival in the United Kingdom: a scoping review. BJC REPORTS 2025; 3:13. [PMID: 40033086 DOI: 10.1038/s44276-025-00126-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/13/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a deadly cancer in the UK despite advancements in curative therapies. Societal conditions and health inequalities influence the development of chronic liver disease and outcomes from complications including HCC. Scoping this emergent evidence-base is required to inform research and solutions for the NHS. METHODS A PRISMA scoping review was performed up to September 2023. Articles exploring health inequalities in HCC involving the UK population were included. RESULTS This review has characterised axes of health inequality and their impact across the HCC care continuum in the UK. Studies predominantly employed a cohort design or population-based analyses, with meta-analyses of surveillance utilisation including only a single UK study. These methodologies provided an appropriate lens to understand longitudinal trends and identify disadvantaged groups. However, important evidence gaps remain, including exploration of patient perspectives, intersectional analyses, and statistical measures of socioeconomic inequity in HCC. CONCLUSIONS HCC is a rapidly growing cause of cancer mortality and disproportionally affects underserved groups, presenting a major public health concern. Further research is required to innovate and evaluate surveillance and management pathways to reduce systemic inequities. Direction is needed at the national level to improve prevention, early diagnosis and access to curative treatment.
Collapse
Affiliation(s)
- Christopher Mysko
- Manchester University NHS Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| | - Stephanie Landi
- Manchester University NHS Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| | - Huw Purssell
- Manchester University NHS Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| | - A Joy Allen
- Roche Diagnostics Limited, Welwyn Garden City, UK
| | - Martin Prince
- Manchester University NHS Foundation Trust, Manchester, UK
| | | | | | | | | | | | | | | | - Neil Hanley
- University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Varinder Singh Athwal
- Manchester University NHS Foundation Trust, Manchester, UK.
- University of Manchester, Manchester, UK.
| |
Collapse
|
42
|
Choi YM, Jang J, Kim DH, Kim Z, Kim E, Choe WH, Kim BJ. PreS1 deletions in genotype C HBV leads to severe hepatic inflammation and hepatocarcinogenesis via the IRE1-JNK axis. JHEP Rep 2025; 7:101274. [PMID: 39980750 PMCID: PMC11840487 DOI: 10.1016/j.jhepr.2024.101274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 02/22/2025] Open
Abstract
Background & Aims Deletion of 15-21 nucleotides covering the preS1 start codon frequently occurs in patients with chronic HBV (CHB) with HBV genotype C and has been reported to be related to progression to hepatocellular carcinoma (HCC). However, the underlying mechanism causing the distinct phenotype of this HBV variant remains largely unknown. We investigated the mechanism by which preS1Del is related to liver disease progression and enhanced HBV replication, focusing on endoplasmic reticulum (ER) stress. Methods The effects of HBV replicative capacity, ER stress signaling, inflammation, cell death, and tumorigenesis resulting from PreS1 deletions were investigated through in vitro and in vivo experiments. Inhibitors of the IRE1-JNK pathway and IL6 blockade were used to examine HCC tumor load induced by preS1 deletions. Results The PreS1Del variant selectively activates the IRE1 pathway, mainly via enhanced colocalization between the ER and HBsAg in infected hepatocytes. This leads to enhanced HBV replication and production of tumor-promoting inflammatory cytokines and IL6 and COX2 via the IRE1-JNK signaling pathway. Furthermore, in vivo data showed that the activation of IRE1-JNK signaling consequently leads to lipid accumulation and apoptosis within 21Del-HBV-infected hepatocytes, collectively driving severe tumorigenesis in the liver. Notably, several inhibitors of the IRE1-JNK pathway dramatically inhibited HBV replication and inflammation induced by 21Del-HBV but not by the wild-type HBV in infected hepatocytes. Furthermore, IL6 blockade significantly reduced HCC tumor load induced by 21Del-HBV. Conclusions PreS1Del leads to enhanced HBV replication and HCC development through IRE1-JNK-IL6/COX2-mediated hepatocyte proliferation and liver inflammation. Inhibitors interfering with the IRE1-JNK-IL6 pathway could selectively inhibit HBV replication and inflammation in preS1Dels, suggesting their potential for the treatment of patients with CHB with preS1-deleted HBV variants. Impact and implications Deletion of 15-21 nucleotides at the preS1 start codon is common in patients with CHB with HBV genotype C and is linked to HCC progression. However, the mechanisms underlying the distinct phenotype of this variant remain largely unknown. We found that the preS1Del variant selectively activates the IRE1 pathway, primarily through enhanced IRE1-JNK-IL6 signaling. Inhibition of either the IRE1-JNK pathway or IL6 reduced HBV replication and tumor load in in vivo HCC models. This study enhances our understanding of the mechanisms of liver disease progression caused by 5' preS1Del variants and provides new insights into treatment strategies for patients with these variants. We believe our findings will resonate with a diverse audience, including patients and their physicians, the medical community, academia, the life sciences sector, and the general public.
Collapse
Affiliation(s)
- Yu-Min Choi
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Junghwa Jang
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Dong Hyun Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Ziyun Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Eunseo Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Won Hyeok Choe
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul 05030, Republic of Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Liver Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Seoul National University Medical Research Center (SNUMRC), Seoul 03080, Republic of Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
43
|
Xu M, Xie P, Liu S, Gao X, Yang S, Hu Z, Zhao Y, Yi Y, Dong Q, Bruns C, Kong X, Hung MC, Ren N, Zhou C. LCAT deficiency promotes hepatocellular carcinoma progression and lenvatinib resistance by promoting triglyceride catabolism and fatty acid oxidation. Cancer Lett 2025; 612:217469. [PMID: 39842501 DOI: 10.1016/j.canlet.2025.217469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Lecithin cholesterol acyltransferase (LCAT), a crucial enzyme in lipid metabolism, plays important yet poorly understood roles in tumours, especially in hepatocellular carcinoma (HCC). In this study, our investigation revealed that LCAT is a key downregulated metabolic gene and an independent risk factor for poor prognosis in patients with HCC. Functional experiments showed that LCAT inhibited HCC cell proliferation, migration and invasion. Mechanistically, LCAT interacts with caveolin-1 (CAV1) to promote the binding of CAV1 to PRKACA and inhibit its phosphorylation, thereby inhibiting triglyceride (TAG) catabolism. On the other hand, LCAT inhibits fatty acid oxidation (FAO) by interacting with CPT1A to promote its ubiquitination and degradation. These events result in an inadequate supply of raw materials and energy and inhibit the malignant behaviours of HCC cells. In addition, LCAT is a reliable predictive biomarker for the efficacy of lenvatinib treatment in HCC patients, and the inhibition of FAO can increase lenvatinib sensitivity in patients with LCATlow HCC. This study revealed that LCAT plays a critical role in the regulation of lipid metabolic reprogramming and is a reliable predictive biomarker for the efficacy of lenvatinib treatment in HCC patients.
Collapse
Affiliation(s)
- Min Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Peiyi Xie
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Shaoqing Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China; Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, P.R. China
| | - Xukang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Shiguang Yang
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, 201199, PR China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201199, PR China; Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201199, PR China
| | - Zhiqiu Hu
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, 201199, PR China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201199, PR China; Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201199, PR China
| | - Yue Zhao
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Yong Yi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China
| | - Qiongzhu Dong
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer of Shanghai Municipal Health Commission, Shanghai, 201199, PR China; Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201199, PR China
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China.
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 40402, Taiwan.
| | - Ning Ren
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China.
| | - Chenhao Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, PR China.
| |
Collapse
|
44
|
Guo R, Wang P. The complex role of regulatory cells in breast cancer: implication for immunopathogenesis and immunotherapy. Breast Cancer 2025; 32:227-241. [PMID: 39589625 DOI: 10.1007/s12282-024-01654-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
Breast cancers (BCs) are frequently linked to an immunosuppressive microenvironment that facilitates tumor evasion of anti-cancer immunity. The cells that suppress the immune system such as regulatory B cells (Bregs), regulatory T cells (Tregs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), play a crucial role in immune resistance. Also, tumor progression and immune evasion of cancers are facilitated by cytokines and factors released by tumor cells or immunosuppressive cells. Targeting these regulatory cells therapeutically, whether through elimination, inactivation, or reprogramming, has resulted in hopeful anti-tumor reactions. Yet, the substantial diversity and adaptability of these cells, both in terms of appearance and function, as well as their variation over time and depending on where they are in the body, have posed significant challenges for using them as reliable biomarkers and creating focused therapies that could target their creation, growth, and various tumor-promoting roles. The immunotherapy approaches in BC and their effectiveness in treating certain subtypes are still in their initial phases. In this review, we thoroughly outlined the characteristics, roles, and possible treatment options for these immune-suppressing cells in the tumor environment.
Collapse
Affiliation(s)
- RuiJuan Guo
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Shandong Province, Yantai City, People's Republic of China
| | - Ping Wang
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Shandong Province, Yantai City, People's Republic of China.
| |
Collapse
|
45
|
Yuan Y, Huang J, Wei G, Hu G, Yu H, Tao Y. Potential mechanism of circKIAA1429 accelerating the progression of hepatocellular carcinoma. Infect Agent Cancer 2025; 20:12. [PMID: 40025575 PMCID: PMC11872318 DOI: 10.1186/s13027-025-00645-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND This study investigates the underlying mechanism of circKIAA1429 (hsa_circ_0084922) in hepatocellular carcinoma (HCC) progression. METHODS circKIAA1429, SETD1A, NAP1L3, and GLIS2 expressions in HCC cells were detected by RT-qPCR or western blot. The stability of circKIAA1429 was tested after treatment with actinomycin D and Rnase R enzyme. circKIAA1429 expression was knocked down, followed by detection of cell proliferation, apoptosis, and migration/invasion using CCK-8, flow cytometry, and transwell. RIP and RNA pull-down were performed to validate the binding between circKIAA1429 and SETD1A, while ChIP analysis determined the enrichment of SETD1A and H3K4me3 or H3K27me3 on GLIS2 or NAP1L3 promoter. A nude mouse xenograft tumor model was establish to test the effect of circKIAA1429 on tumorigenicity. RESULTS circKIAA1429 and NAP1L3 were highly expressed in HCC cells, while GLIS2 was poorly expressed. Knockdown of circKIAA1429 repressed cell proliferation/invasion/migration and facilitated apoptosis. Mechanistically, circKIAA1429 directly interacted with SETD1A to reduce the enrichment of SETD1A and H3K4me3 or H3K27me3 on GLIS2 or NAP1L3 promoter, thus diminishing GLIS2 expression and elevating NAP1L3 expression. In vivo, circKIAA1429 promotes tumorigenesis via GLIS2/NAP1L3. CONCLUSION circKIAA1429 interacts with SETD1A to inhibit the enrichment of H3K4me3 and H3K27me3 on GLIS2 or NAP1L3 promoter, thus inhibiting/promoting the expression of GLIS2/NAP1L3 and accelerating the progression of HCC.
Collapse
Affiliation(s)
- Yiting Yuan
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Junwei Huang
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Guifen Wei
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Guang Hu
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Hongmei Yu
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Yiming Tao
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China.
| |
Collapse
|
46
|
Brahmania M, Congly S, Sachar Y, Burak KW, Lethebe B, Szostakiwskyj JH, Lautner D, Medellin A, Bhayana D, Wong J, Nguyen H, Sadler MD, Borman M, Aspinall AI, Coffin CS, Swain M, Shaheen A. Dedicated Automatic Recall Hepatocellular Cancer Surveillance Programme Demonstrates High Retention: A Population-Based Cohort Study. Liver Int 2025; 45:e70020. [PMID: 39927626 PMCID: PMC11809127 DOI: 10.1111/liv.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/28/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION Patient, clinician, and system-related barriers may affect adherence to hepatocellular carcinoma (HCC) surveillance programmes. The impact of a dedicated automated recall HCC surveillance programme on retention rates in patients eligible for screening is unknown. We aimed to describe and evaluate a large HCC surveillance programme in a publicly funded healthcare system. METHODS Data were collected from January 1, 2013, to December 31, 2022, from a retrospective cohort of subjects enrolled in a publicly funded automated recall semi-annual surveillance programme as per the American Association for the Study of Liver Disease HCC guidance in the Calgary Health Zone (~1.6 million), Canada. Patients were excluded if there was incomplete data or did not meet indications for surveillance. Cox regression was used to identify predictors of non-retention to surveillance. RESULTS A total of 7269 patients were included. The median was age 55.5 years (IQR: 45.5-63.8), 60% were male, 46% were of Asian descent, 51% had HBV infection, and 36% had cirrhosis (35% alcohol-related). Median follow-up was 4.9 years (IQR: 1.5-7.2). Overall, 52% (n = 3768) of patients were retained in the surveillance programme, while 8.3% (n = 603) left for potential medical reasons, and 40% (n = 2898) were lost in follow-up. The median time in the programme for those lost in follow-up was 0.81 years (IQR: 0.0-2.8) compared to 6.75 years if retained (IQR: 5.6-8.6; p < 0.001). In multivariable Cox regression analysis, HCV aetiology (HR 1.41; CI 1.23-1.62, p < 0.01), African ethnicity (HR 1.20, CI 1.02-1.42, p = 0.03), and cirrhosis (HR 1.16, CI 1.05-1.28, p < 0.01) increased risk of dropout. On interaction analysis, Hepatitis B amongst cirrhotic patients also increased risk of dropout (HR 1.48, CI 1.05-2.07, p = 0.02). CONCLUSION A dedicated automated recall HCC surveillance programme has a high retention rate in a large multi-ethnic cohort of patients while identifying certain marginalised patient populations, such as those with viral liver disease, cirrhosis, or African ethnicity, as particularly vulnerable to loss to follow-up.
Collapse
Affiliation(s)
- Mayur Brahmania
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
- O'Brien Institute of Public HealthSchulich School of MedicineLondonOntarioCanada
| | - Stephen Congly
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
- O'Brien Institute of Public HealthSchulich School of MedicineLondonOntarioCanada
| | - Yashasavi Sachar
- Division of Internal Medicine, Department of MedicineSchulich School of MedicineLondonOntarioCanada
| | - Kelly W. Burak
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
- Department of OncologyCumming School of MedicineCalgaryAlbertaCanada
| | - Brendan Lethebe
- Clinical Research UnitCumming School of Medicine, University of CalgaryCalgaryAlbertaCanada
| | | | - David Lautner
- Department of RadiologyCumming School of Medicine, University of CalgaryCalgaryAlbertaCanada
| | - Alexandra Medellin
- Department of RadiologyCumming School of Medicine, University of CalgaryCalgaryAlbertaCanada
| | - Deepak Bhayana
- Department of RadiologyCumming School of Medicine, University of CalgaryCalgaryAlbertaCanada
| | - Jason Wong
- Department of RadiologyCumming School of Medicine, University of CalgaryCalgaryAlbertaCanada
| | - Henry Nguyen
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
| | - Matthew D. Sadler
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
| | - Meredith Borman
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
| | - Alexander I. Aspinall
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
| | - Carla S. Coffin
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
| | - Mark Swain
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
| | - Abdel‐Aziz Shaheen
- Division of Gastroenterology and Hepatology, Department of MedicineSchulich School of MedicineLondonOntarioCanada
- O'Brien Institute of Public HealthSchulich School of MedicineLondonOntarioCanada
| |
Collapse
|
47
|
Li SB, Zhao TSY, Ye Z, Zou J, Yuan X, Zhou XL, Liang CQ, Li KZ, Huang LZ. Antitumor effects of BPCO on liver cancer cells. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-13. [PMID: 39985778 DOI: 10.1080/10286020.2025.2467318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
Esculetin is a coumarin compound with anticancer, antioxidant, and anti-inflammatory activities. In this study, we synthesized an esculetin derivative, 6,7-bis(Pentyloxy)-2H-Chromen-2-One (BPCO), through etherification. BPCO inhibited the proliferation of HepG2 cells in a dose- and time-dependent manner. It also inhibited cell migration, promoted apoptosis, and caused cell cycle arrest at the G1 phase. Additionally, BPCO downregulated the expression levels of Bcl-2 and Bcl-XL and upregulated the expression levels of Bax and Bak. This study shows that BPCO inhibits hepatocellular carcinoma cell proliferation and induces apoptosis, providing a basis for further study of BPCO as an antitumor agent.
Collapse
Affiliation(s)
- Shan-Bin Li
- College of Biotechnology, Guilin Medical University, Guilin541199, China
- Key Laboratory of Molecular Medical Engineering, Education Department of Guangxi Zhuang Autonomous Region, Guilin541199, China
| | - Tong-Shi-Yao Zhao
- College of Biotechnology, Guilin Medical University, Guilin541199, China
| | - Zhen Ye
- College of Pharmacy, Guilin Medical University, Guilin541199, China
| | - Jian Zou
- College of Biotechnology, Guilin Medical University, Guilin541199, China
| | - Xi Yuan
- College of Biotechnology, Guilin Medical University, Guilin541199, China
| | - Xian-Li Zhou
- College of Biotechnology, Guilin Medical University, Guilin541199, China
- Key Laboratory of Molecular Medical Engineering, Education Department of Guangxi Zhuang Autonomous Region, Guilin541199, China
| | - Cheng-Qin Liang
- College of Pharmacy, Guilin Medical University, Guilin541199, China
| | - Kang-Zhi Li
- College of Biotechnology, Guilin Medical University, Guilin541199, China
| | - Lan-Zhen Huang
- Science Experiment Center, Guilin Medical University, Guilin541199, China
| |
Collapse
|
48
|
Xie X, Tsui YM, Zhang VX, Yu TCY, Husain A, Chiu YT, Tian L, Lee E, Lee JMF, Ma HT, Ho DWH, Sze KMF, Ng IOL. Nuclear localization of BRCA1-associated protein 1 is important in suppressing hepatocellular carcinoma metastasis via CTCF and NRF1/OGT axis. Cell Death Dis 2025; 16:123. [PMID: 39984455 PMCID: PMC11845619 DOI: 10.1038/s41419-025-07451-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 01/31/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
Germline mutations of the deubiquitinase BRCA1-associated protein 1 (BAP1) lead to the "BAP1 cancer syndrome" characterized by development of cancers. However, the role of BAP1 in hepatocellular carcinoma (HCC) is unclear. We found that BAP1 was upregulated at mRNA level in human HCCs and significantly correlated with a more aggressive tumour behaviour. Intriguingly, we observed cytoplasmic but no or minimal nuclear BAP1 in human HCC samples by immunohistochemistry. We observed that, while BAP1 protein was found mainly in the cytoplasm and less in the nuclei of HCC cell lines, BAP1 expression was predominantly nuclear in HepG2 cells, by cell fractionation and immunofluorescence analyses. Functionally, in the orthotopic liver injection mouse model, silencing the BAP1 predominant nuclear expression of HepG2 cells promoted intrahepatic tumor metastasis, with more frequent tumor microsatellite formation and venous invasion. With transcriptomic profiling, we identified RHOJ amongst the downregulated targets in HepG2 cells upon BAP1 knockdown. Subsequent overexpression of RHOJ suppressed cell migration in HCC cells, suggesting that BAP1 might upregulate RHOJ resulting in reduced cell migratory ability of HCC cells. Furthermore, we identified two transcription factors, CTCF and NRF1, which activated BAP1 transcription by binding to BAP1 promoter region. On the other hand, we uncovered that O-linked N-acetylglucosamine (GlcNAc) transferase (OGT) physically bound to BAP1 in the nucleus, resulting in diminished stability of the nuclear BAP1. Intriguingly, OGT transcription was upregulated and was also under the control of CTCF and NRF1 in human HCC, acting as a negative regulator of BAP1. To summarize, this study uncovered the underlying mechanisms of the regulation of BAP1 and that loss of the nuclear localization of BAP1 protein contributed to enhanced cell migration in vitro and more aggressive tumor behavior in human HCCs.
Collapse
Affiliation(s)
- Xiaoyu Xie
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yu-Man Tsui
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Vanilla Xin Zhang
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Tiffany Ching-Yun Yu
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Abdullah Husain
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yung-Tuen Chiu
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Lu Tian
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Eva Lee
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Joyce Man-Fong Lee
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hoi-Tang Ma
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Daniel Wai-Hung Ho
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Karen Man-Fong Sze
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong.
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.
| | - Irene Oi-Lin Ng
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong.
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
49
|
Zhang Z, Yu G, Eresen A, Hou Q, Webster S, Amirrad F, Nauli S, Zhang Z. Therapy Combining Sorafenib and Natural Killer Cells for Hepatocellular Carcinoma: Insights from Magnetic Resonance Imaging and Histological Analyses. Cancers (Basel) 2025; 17:699. [PMID: 40002292 PMCID: PMC11853247 DOI: 10.3390/cancers17040699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Hepatocellular carcinoma (HCC) remains a significant global health issue due to its high mortality rate and resistance to standard treatments. Sorafenib, the first-line systemic therapy for unresectable HCC, shows limited effectiveness due to resistance and severe side effects. Recent studies suggest that combining sorafenib with immunotherapy, particularly natural killer (NK) cells, may improve treatment outcomes. Methods: This study examined the effectiveness of sorafenib combined with NK cells pretreated with interleukin-12 (IL-12) and interleukin-18 (IL-18) in a rat HCC model. Tumor progression and treatment outcomes were assessed using MRI and histological analysis. Results: The results show that combination therapy significantly reduced tumor growth, increased tumor cell density, and inhibited angiogenesis and fibrosis in the tumor microenvironment. The sorafenib- and IL-12/IL-18-pretreated NK cell combination enhanced tumor inhibition by overcoming drug resistance and modulating the immune response. Conclusions: This study suggests that this combination therapy could be a promising strategy for treating HCC, offering both direct antitumor effects and modification of the tumor microenvironment for better clinical outcomes.
Collapse
Affiliation(s)
- Zigeng Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA 92617, USA; (A.E.); (Q.H.)
| | - Guangbo Yu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA;
| | - Aydin Eresen
- Department of Radiological Sciences, University of California Irvine, Irvine, CA 92617, USA; (A.E.); (Q.H.)
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92617, USA
| | - Qiaoming Hou
- Department of Radiological Sciences, University of California Irvine, Irvine, CA 92617, USA; (A.E.); (Q.H.)
| | - Sha Webster
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA 92618, USA; (S.W.); (F.A.); (S.N.)
| | - Farideh Amirrad
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA 92618, USA; (S.W.); (F.A.); (S.N.)
| | - Surya Nauli
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA 92618, USA; (S.W.); (F.A.); (S.N.)
- Department of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA 92617, USA; (A.E.); (Q.H.)
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA;
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92617, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA 92617, USA
| |
Collapse
|
50
|
He Y, Li H, Shi Q, Liu Y, Pan Q, He X. The liver-specific long noncoding RNA FAM99B inhibits ribosome biogenesis and cancer progression through cleavage of dead-box Helicase 21. Cell Death Dis 2025; 16:97. [PMID: 39952918 PMCID: PMC11829061 DOI: 10.1038/s41419-025-07401-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/09/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
Emerging evidence has demonstrated that long noncoding RNAs (lncRNAs) are promising targets or agents for the treatment of human cancers. Most liver-specific lncRNAs exhibit loss of expression and act as tumor suppressors in liver cancer. Modulating the expression of these liver-specific lncRNAs is a potential approach for lncRNA-based gene therapy for hepatocellular carcinoma (HCC). Here, we report that the expression of the liver-specific lncRNA FAM99B is significantly decreased in HCC tissues and that FAM99B suppresses HCC cell proliferation and metastasis both in vitro and in vivo. FAM99B promotes the nuclear export of DDX21 through XPO1, leading to further cleavage of DDX21 by caspase3/6 in the cytoplasm. FAM99B inhibits ribosome biogenesis by inhibiting ribosomal RNA (rRNA) processing and RPS29/RPL38 transcription, thereby reducing global protein synthesis through downregulation of DDX21 in HCC cells. Interestingly, the FAM99B65-146 truncation exhibits tumor-suppressive effects in vivo and in vitro. Moreover, GalNAc-conjugated FAM99B65-146 inhibits the growth and metastasis of orthotopic HCC xenografts, providing a new strategy for the treatment of HCC. This is the first report of the use of a lncRNA as an agent rather than a target in tumor treatment. Graphical illustration of the mechanism of FAM99B in HCC.
Collapse
MESH Headings
- Humans
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- DEAD-box RNA Helicases/metabolism
- DEAD-box RNA Helicases/genetics
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Animals
- Ribosomes/metabolism
- Ribosomes/genetics
- Cell Proliferation/genetics
- Cell Line, Tumor
- Mice
- Karyopherins/metabolism
- Karyopherins/genetics
- Disease Progression
- Mice, Nude
- Gene Expression Regulation, Neoplastic
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Exportin 1 Protein
- Mice, Inbred BALB C
- Liver/metabolism
- Liver/pathology
- Male
- RNA, Ribosomal/metabolism
- RNA, Ribosomal/genetics
Collapse
Affiliation(s)
- Yifei He
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hongquan Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Qili Shi
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yanfang Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qiaochu Pan
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xianghuo He
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|